WO2013008095A1 - Novel pyrrolo pyrimidine derivatives - Google Patents

Novel pyrrolo pyrimidine derivatives Download PDF

Info

Publication number
WO2013008095A1
WO2013008095A1 PCT/IB2012/001699 IB2012001699W WO2013008095A1 WO 2013008095 A1 WO2013008095 A1 WO 2013008095A1 IB 2012001699 W IB2012001699 W IB 2012001699W WO 2013008095 A1 WO2013008095 A1 WO 2013008095A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
phenyl
fluoro
pyrrolo
pyrimidin
Prior art date
Application number
PCT/IB2012/001699
Other languages
French (fr)
Inventor
Richard Heng
Elizabeth Kate HOEGENAUER
Guido Koch
Robert Alexander Pulz
Anna Vulpetti
Rudolf Waelchli
Original Assignee
Novartis Ag
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Novartis Ag filed Critical Novartis Ag
Priority to ES12761655.5T priority Critical patent/ES2548414T3/en
Priority to EA201490229A priority patent/EA201490229A1/en
Priority to JP2014517980A priority patent/JP6145451B2/en
Priority to KR1020147002986A priority patent/KR20140058543A/en
Priority to EP12761655.5A priority patent/EP2729466B1/en
Priority to AU2012282229A priority patent/AU2012282229B2/en
Priority to CN201280033935.9A priority patent/CN103732596B/en
Priority to MX2014000338A priority patent/MX2014000338A/en
Priority to CA2841111A priority patent/CA2841111A1/en
Priority to US14/130,536 priority patent/US9233111B2/en
Priority to BR112014000314A priority patent/BR112014000314A2/en
Publication of WO2013008095A1 publication Critical patent/WO2013008095A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the present invention describes new pyrrolo pyrimidine derivatives that are good drug candidates.
  • the compounds of the present invention may generally exhibit a selective inhibition of Bruton's tyrosine kinase (Btk).
  • Btk-deficient mice are protected in standard preclinical models for rheumatoid arthritis (Jansson and Holmdahl, 1993), systemic lupus erythematosus (Steinberg, B.J. et al., J. Clin. Invest, 70, 587-597, 1982), as well as allergic disease and anaphylaxis (Hata.D. et al., J. Exp. Med. 187, 1235-1247, 1998).
  • many cancers and lymphomas express Btk and appear to be dependent on Btk function (Davis, R.E. et al., Nature, 463, 88-92, 2010).
  • inhibition of Btk activity may be useful in the treatment of immune disorders such as rheumatoid arthritis, systemic lupus erythematosus, allergic diseases, anaphylaxis and inflammatory conditions.
  • inhibition of Btk may be useful in the treatment of cancers of haematopoietic origin including chronic myelogenous leukemia, myeloid leukemia, non-Hodgkin lymphoma and other B cell lymphomas.
  • the compounds of the present invention may therefore potentially be useful in the treatment of a wide range of disorders, particularly Btk-related diseases or disorders, and may for example be useful in the treatment of autoimmune disorders, inflammatory diseases, allergic diseases, airway diseases, such as asthma and chronic obstructive pulmonary disease (COPD), transplant rejection, or cancers e.g. of hematopoietic origin or solid tumors. More particularly, the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof;
  • R1 is hydrogen, CrC 6 alkyi optionally substituted by hydroxy
  • R2 is hydrogen or halogen
  • R3 is hydrogen or halogen
  • R4 is hydrogen
  • R5 is phenyl optionally substituted by halogen; SF 5 ; NR6R7; hydroxy; C C 6 alkoxy; d- C 6 alkenyl; CrC 6 alkyi carbonyl; CrC 6 alkyi optionally substituted by hydroxy, halogen, or Ci-C 6 alkoxy; or C 3 -C 6 cycloalkyi optionally substituted by halogen, hydroxy, or C C 6 alkyi optionally substituted by halogen; or
  • R5 is a 4 - 14 membered mono- or bicyclic heterocyclyl or heteroaryl ring system comprising 1 , 2 or 3 heteroatoms selected from N, S and O that ring being optionally substituted by halogen; hydroxy; CrC 6 alkoxy optionally substituted by hydroxy or halogen; or CrC 6 alkyi optionally substituted by hydroxy or halogen;
  • R4 and R5 together with the atoms to which they are bound form a piperidone ring, optionally comprising an annulated phenyl ring, any such ring being optionally substituted by C C 6 alkyi, C C 6 alkoxy, or C 3 -C 6 cycloalkyi each of which substitution member may optionally be substituted by halogen or hydroxy;
  • R6 and R7 are independently selected from hydrogen or CrC 6 alkyi
  • R6 and R7 together with the nitrogen atom to which they are bound form a 4 - 8 membered saturated azacycloalkane ring, optionally substituted by halogen, hydroxy or
  • X is O, S(0) n wherein n is 0, 1 or 2, or wherein q is 2 or 3, and R10 is absent; or X is CH or N; and R10 is hydrogen, hydroxy, -NR6R7, -CO-R1 1 , -S(0) p -R12 wherein p is 1 or 2,
  • R 1 1 is Ci-C 6 alkyl optionally substituted by hydroxy, cyano, halogen, carboxy or CrC 6 alkoxy carbonyloxy; or NR6R7; and
  • R12 is Ci-C 6 alkyl or NR6R7.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
  • R2 and R3 are independently hydrogen or fluoro
  • R4 is hydrogen
  • R5 is phenyl substituted by halogen
  • CrC 6 alkoxy or CrC 6 alkyl optionally substituted by halogen or hydroxy
  • C 3 -C 6 cycloalkyi optionally substituted by halogen, hydroxy, C C 6 substituted by halogen
  • X is O, S(0) n wherein n is 0, 1 or 2, or wherein q is 2 or 3, and R10 is absent;
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
  • R2 and R3 are independently hydrogen or fluoro
  • R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one optionally substituted by C 3 -C 6 cycloalkyi or C C 6 substituted by hydroxy
  • X is O, S(0) n wherein n is 0, 1 or 2, or wherein q is 2 or 3, and R10 is absent; and the remaining variables are as defined above.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
  • R2 and R3 are independently hydrogen or fluoro
  • R4 is hydrogen
  • R5 is phenyl substituted by halogen
  • CrC 6 alkoxy CrC 6 alkyl optionally substituted by halogen or hydroxy
  • C 3 -C 6 cycloalkyi optionally substituted by halogen, hydroxy, or d- C 6 alkyl optionally substituted by halogen
  • X stands for O and R10 is absent
  • X stands for N
  • R10 is hydrogen or -CO-R1 1 , and the remaining variables are as defined above.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
  • R2 and R3 are independently hydrogen or fluoro
  • R4 is hydrogen
  • R5 is phenyl substituted by CrC 6 alkoxy; CrC 6 alkyl optionally substituted by halogen or hydroxy; or C 3 -C 6 cycloalkyl optionally substituted by halogen, hydroxy, or CrC 6 alkyl optionally substituted by halogen
  • X stands for N
  • R10 is hydrogen or -CO-R1 1
  • R1 1 stands for NR6R7 wherein R6 and R7 are independently hydrogen or methyl; and the remaining variables are as defined above.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
  • R2 and R3 are independently hydrogen or fluoro
  • R4 is hydrogen
  • R5 is azetidine optionally substituted by CrC 6 alkoxy
  • X stands for N
  • R10 is hydrogen or -CO-R1 1 , and the remaining variables are as defined above.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
  • R2 and R3 are independently hydrogen or fluoro
  • R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one optionally substituted by C 3 -C 6 cycloalkyl or C C 6 alkyl optionally substituted by hydroxy
  • X stands for N
  • R10 is hydrogen or -CO-R1 1 , and the remaining variables are as defined above.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
  • R2 and R3 are independently hydrogen or fluoro
  • R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one substituted by C 3 -C 6 cycloalkyl or C C 6 alkyl optionally substituted by hydroxy in the 6-position of said isoquinolin-ring
  • X stands for N
  • R10 is hydrogen or -CO-R1 1 , and the remaining variables are as defined above.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
  • R2 and R3 are independently hydrogen or fluoro
  • R4 is hydrogen
  • R5 is azetidine optionally substituted by CrC 6 alkoxy
  • X stands for O, and the remaining variables are as defined above.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
  • R2 and R3 are independently hydrogen or fluoro
  • R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one optionally substituted by C 3 -C 6 cycloalkyi or C C 6 alkyl optionally substituted by hydroxy
  • X stands for O, and the remaining variables are as defined above.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
  • R2 and R3 are independently hydrogen or fluoro
  • R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one substituted by C 3 -C 6 cycloalkyi or C C 6 alkyl optionally substituted by hydroxy in the 6-position of said isoquinolin-ring
  • X stands for O, and the remaining variables are as defined above.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
  • R2 and R3 are independently from each other selected from hydrogen and halogen
  • R4 is hydrogen
  • R5 is phenyl substituted one or more times by halogen
  • X stands for O or S
  • R10 is absent.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is methyl or hydroxymethyl , R2 and R3 are independently from each other selected from hydrogen and halogen, R4 is hydrogen, R5 is phenyl substituted one or more times by halogen, C 3 -C 6 cycloalkyi, or Ci-C 6 alkyl optionally substituted by hydroxy, X stands for N, R10 is hydrogen or -CO-R1 1 , and R1 1 is NR6R7 wherein R6 and R7 are independently selected from C Ce-alkyl.
  • R1 is methyl or hydroxymethyl
  • R2 and R3 are independently from each other selected from hydrogen and halogen
  • R4 is hydrogen
  • R5 is phenyl substituted one or more times by halogen, C 3 -C 6 cycloalkyi, or Ci-C 6 alkyl optionally substituted by hydroxy
  • X stands for N
  • R10 is hydrogen or -CO-R
  • R1 is hydrogen, methyl or hydroxymethyl
  • R1 is methyl or hydroxymethyl
  • R2 and R3 are independently hydrogen or fluoro
  • R1 is methyl or hydroxymethyl and R2 and R3 are independently hydrogen or fluoro;
  • R4 is hydrogen
  • R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one optionally substituted by C 3 -C 6 cycloalkyi or CrC 6 alkyl optionally substituted by hydroxy;
  • R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one optionally substituted by C 3 -C 6 cycloalkyi or CrC 6 alkyl optionally substituted by hydroxy in the 6- position of the 3,4-dihydro-2H-isoquinolin-1-one ring;
  • R5 is phenyl optionally substituted by -NR6R7, halogen; CrC 6 alkoxy, CrC 6 alkenyl, C 3 -C 6 cycloalkyi, or CrC 6 alkyl optionally substituted by halogen or hydroxy;
  • R5 is phenyl substituted by -NR6R7, halogen, CrC 6 alkoxy or C C 6 alkyl optionally substituted by halogen or hydroxy;
  • R5 is phenyl substituted by halogen, CrC 6 alkoxy, C 3 -C 6 cycloalkyi or C C 6 alkyl optionally substituted by fluoro or hydroxy;
  • R5 is a 4-, 5-, 6-, or 7-membered monocyclic heterocycle, or a 7-, 8-, 9-, 10-, 1 1-, or 12-membered bicyclic heterocycle comprising 1 , 2 or 3 heteroatoms selected from N, S and O that ring being optionally substituted by halogen; hydroxy; CrC 6 alkoxy optionally substituted by hydroxy or halogen; or C C 6 alkyl optionally substituted by hydroxy or halogen;
  • R5 is a 4-, 5-, 6-, or 7-membered monocyclic heterocycle comprising 1 or 2 heteroatoms selected from N, S and O that ring being optionally substituted by halogen; hydroxy; CrC 6 alkoxy optionally substituted by hydroxy or halogen; or Ci-C 6 alkyl optionally substituted by hydroxy or halogen;
  • R5 is azetidine substituted by C C 6 alkoxy or C C 6 alkyl; 14.
  • X is O and R10 is absent or is N and R10 is H or CO-R1 1 ;
  • X is O and R10 is absent or is N and R10 is CO-R1 1 ;
  • X is N and R10 is CO-R11 ;
  • R11 is NR6R7 and R6 and R7 are independently selected from hydrogen or d- C 6 alkyl;
  • R11 is NR6R7 and R6 and R7 are independently selected from d-C 6 alkyl;
  • R11 is NR6R7 and R6 and R7 are independently selected from C C 3 alkyl;
  • R11 is NR6R7 and R6 and R7 are methyl.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof for use as a medicament.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of a disease or disorder mediated by Btk.
  • the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof, which is selected from:
  • alkyl refers to a fully saturated branched or unbranched hydrocarbon moiety having up to 20 carbon atoms. Unless otherwise provided, alkyl refers to hydrocarbon moieties having 1 to 16 carbon atoms, 1 to 10 carbon atoms, 1 to 7 carbon atoms, or 1 to 4 carbon atoms.
  • alkyl include, but are not limited to, methyl, ethyl, n-propyl, / ' so-propyl, n-butyl, sec-butyl, / ' so-butyl, tert- butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, 3-methylhexyl, 2,2- dimethylpentyl, 2,3- dimethylpentyl, n-heptyl, n-octyl, n-nonyl, n-decyl and the like.
  • alkenyl refers to an unsaturated branched or unbranched hydrocarbon moiety having 2 to 20 carbon atoms. It comprises 2 to 20 carbon atoms, Unless otherwise provided, alkenyl refers to moieties having 2 to 16 carbon atoms, 2 to 10 carbon atoms, 2 to 7 carbon atoms, or 2 to 4 carbon atoms.
  • alkenyl include, but are not limited to, ethenyl, n-propenyl, / ' so-propenyl, n- butenyl, sec-butenyl, / ' so-butenyl, fert-butenyl, n-pentenyl, isopentenyl, neopentenyl, n- hexenyl, 3-methylhexenyl, 2,2- dimethylpentenyl, 2,3-dimethylpentenyl, n-heptenyl, n- octenyl, n-nonenyl, n-decenyl and the like.
  • alkoxy refers to alkyl-O-, wherein alkyl is defined herein above.
  • Representative examples of alkoxy include, but are not limited to, methoxy, ethoxy, propoxy, 2-propoxy, butoxy, fert-butoxy, pentyloxy, hexyloxy, cyclopropyloxy-, cyclohexyloxy- and the like.
  • alkoxy groups typically have about 1-7, more preferably about 1-4 carbons.
  • cycloalkyl refers to saturated or unsaturated monocyclic, bicyclic or tricyclic hydrocarbon groups of 3-12 carbon atoms. Unless otherwise provided, cycloalkyl refers to cyclic hydrocarbon groups having between 3 and 9 ring carbon atoms or between 3 and 7 ring carbon atoms. Exemplary monocyclic hydrocarbon groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl and cyclohexenyl and the like.
  • Exemplary bicyclic hydrocarbon groups include bornyl, indyl, hexahydroindyl, tetrahydronaphthyl, decahydronaphthyl, bicyclo[2.1.1]hexyl, bicyclo[2.2.1]heptyl, bicyclo[2.2.1]heptenyl, 6,6- dimethylbicyclo[3.1.1]heptyl, 2,6,6-trimethylbicyclo[3.1.1]heptyl, bicyclo[2.2.2]octyl and the like.
  • Exemplary tricyclic hydrocarbon groups include adamantyl and the like.
  • azacycloalkane refers to saturated or unsaturated monocyclic, bicyclic or tricyclic hydrocarbon groups of 3-12 carbon atoms as defined for “cycloalkyl", wherein one carbon atom is replaced by a nitrogen atom.
  • azacycloalkyi refers to cyclic aza-hydrocarbon groups having between 2 and 9 ring carbon atoms and one nitrogen atom or between 2 and 7 ring carbon atoms and one nitrogen atom.
  • Exemplary monocyclic aza-hydrocarbon groups include, but are not limited to, aziridinyl, azetidinly, pyrollidinyl, piperidinyl, azepanyl, dihydroazepinyl and the like.
  • halogen refers to fluoro, chloro, bromo, and iodo.
  • heterocyclic may refer to a saturated or unsaturated non-aromatic ring or ring system, e.g., which is a 4-, 5-, 6-, or 7-membered monocyclic, 7-, 8-, 9-, 10-, 1 1-, or 12-membered bicyclic or 10-, 1 1-, 12-, 13-, 14- or 15-membered tricyclic ring system and contains at least one heteroatom selected from O, S and N, where the N and S can also optionally be oxidized to various oxidation states.
  • the heterocyclic group can be attached at a heteroatom or a carbon atom.
  • the heterocyclyl may include fused or bridged rings as well as spirocyclic rings.
  • heterocycles include azetidine, tetrahydrofuran (THF), dihydrofuran, 1 , 4- dioxane, morpholine, 1 ,4-dithiane, piperazine, piperidine, 1 ,3-dioxolane, imidazolidine, imidazoline, pyrroline, pyrrolidine, tetrahydropyran, dihydropyran, oxathiolane, dithiolane, 1 ,3-dioxane, 1 ,3-dithiane, oxathiane, thiomorpholine, and the like.
  • aryloxy refers to both an -O-aryl and an -O-heteroaryl group, wherein aryl and heteroaryl are defined herein.
  • heteroaryl refers to a 5-14 membered monocyclic- or bicyclic- or tricyclic-aromatic ring system, having 1 to 8 heteroatoms selected from N, O or S.
  • the heteroaryl is a 5-10 membered ring system (e.g., 5-7 membered monocycle or an 8-10 memberred bicycle) or a 5-7 membered ring system.
  • Typical heteroaryl groups include 2- or 3-thienyl, 2- or 3-furyl, 2- or 3-pyrrolyl, 2-, 4-, or 5- imidazolyl, 3-, 4-, or 5- pyrazolyl, 2-, 4-, or 5-thiazolyl, 3-, 4-, or 5-isothiazolyl, 2-, 4-, or 5- oxazolyl, 3-, 4-, or 5-isoxazolyl, 3- or 5-1 ,2,4-triazolyl, 4- or 5-1 ,2, 3-triazolyl, tetrazolyl, 2- , 3-, or 4-pyridyl, 3- or 4-pyridazinyl, 3-, 4-, or 5-pyrazinyl, 2-pyrazinyl, and 2-, 4-, or 5- pyrimidinyl.
  • heteroaryl also refers to a group in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where the radical or point of attachment is on the heteroaromatic ring.
  • Nonlimiting examples include 1-, 2-, 3-, 5-, 6-, 7-, or 8- indolizinyl, 1-, 3-, 4-, 5-, 6-, or 7-isoindolyl, 2-, 3-, 4-, 5-, 6-, or 7-indolyl, 2-, 3- , 4-, 5-, 6-, or 7-indazolyl, 2-, 4-, 5-, 6-, 7-, or 8- purinyl, 1-, 2-, 3-, 4-, 6-, 7-, 8-, or 9- quinolizinyl, 2-, 3-, 4-, 5-, 6-, 7-, or 8-quinoliyl, 1-, 3-, 4-, 5-, 6-, 7-, or 8-isoquinoliyl, 1-, 4-, 5-, 6-, 7-, or 8-phthalazinyl, 2-, 3-, 4-, 5-, or 6-naphthyridinyl, 2-, 3-, 4-, 5-, or 6-naphthyridinyl, 2-, 3- , 5-, 6-, 7-, or 8
  • Typical fused heteroary groups include, but are not limited to 2-, 3-, 4-, 5-, 6-, 7-, or 8-quinolinyl, 1-, 3-, 4-, 5-, 6-, 7-, or 8-isoquinolinyl, 2-, 3-, 4-, 5-, 6-, or 7-indolyl, 2-, 3-, 4-, 5-, 6-, or 7-benzo[b]thienyl, 2-, 4-, 5- , 6-, or 7- benzoxazolyl, 2-, 4-, 5-, 6-, or 7-benzimidazolyl, and 2-, 4-, 5-, 6-, or 7-benzothiazolyl.
  • salt refers to an acid addition or base addition salt of a compound of the invention.
  • Salts include in particular “pharmaceutical acceptable salts”.
  • pharmaceutically acceptable salts refers to salts that retain the biological effectiveness and properties of the compounds of this invention and, which typically are not biologically or otherwise undesirable.
  • the compounds of the present invention are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto.
  • Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids, e.g., acetate, aspartate, benzoate, besylate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlortheophyllonate, citrate, ethandisulfonate, fumarate, gluceptate, gluconate, glucuronate, hippurate, hydroiodide/iodide, isethionate, lactate, lactobionate, laurylsulfate, malate, maleate, malonate, mandelate, mesylate, methylsulphate, naphthoate, napsylate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen
  • Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, sulfosalicylic acid, and the like.
  • Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
  • Inorganic bases from which salts can be derived include, for example, ammonium salts and metals from columns I to XII of the periodic table.
  • the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
  • Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like.
  • Certain organic amines include isopropylamine, benzathine, cholinate, diethanolamine, diethylamine, lysine, meglumine, piperazine and tromethamine.
  • the pharmaceutically acceptable salts of the present invention can be synthesized from a basic or acidic moiety, by conventional chemical methods.
  • such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid.
  • a stoichiometric amount of the appropriate base such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like
  • Such reactions are typically carried out in water or in an organic solvent, or in a mixture of the two.
  • use of non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile is desirable, where practicable.
  • any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds.
  • Isotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 18 F 31 P, 32 P, 35 S, 36 CI, 125 l respectively.
  • the invention includes various isotopically labeled compounds as defined herein, for example those into which radioactive isotopes, such as 3 H and 14 C, or those into which non-radioactive isotopes, such as 2 H and 13 C are present.
  • isotopically labelled compounds are useful in metabolic studies (with 14 C), reaction kinetic studies (with, for example 2 H or 3 H), detection or imaging techniques, such as positron emission tomography (PET) or single- photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients.
  • PET positron emission tomography
  • SPECT single- photon emission computed tomography
  • an 18 F or labeled compound may be particularly desirable for PET or SPECT studies.
  • Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagents in place of the non-labeled reagent previously employed.
  • isotopic enrichment factor means the ratio between the isotopic abundance and the natural abundance of a specified isotope.
  • a substituent in a compound of this invention is denoted deuterium, such compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation).
  • solvates in accordance with the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D 2 0, de- acetone, d 6 -DMSO.
  • Compounds of the invention i.e. compounds of formula (I) that contain groups capable of acting as donors and/or acceptors for hydrogen bonds may be capable of forming co- crystals with suitable co-crystal formers.
  • These co-crystals may be prepared from compounds of formula (I) by known co-crystal forming procedures. Such procedures include grinding, heating, co-subliming, co-melting, or contacting in solution compounds of formula (I) with the co-crystal former under crystallization conditions and isolating co- crystals thereby formed.
  • Suitable co-crystal formers include those described in WO 2004/078163.
  • the invention further provides co-crystals comprising a compound of formula (I).
  • the term "pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drug stabilizers, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, and the like and combinations thereof, as would be known to those skilled in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, pp. 1289- 1329).
  • a therapeutically effective amount of a compound of the present invention refers to an amount of the compound of the present invention that will elicit the biological or medical response of a subject, for example, reduction or inhibition of an enzyme or a protein activity, or ameliorate symptoms, alleviate conditions, slow or delay disease progression, or prevent a disease, etc.
  • a therapeutically effective amount refers to the amount of the compound of the present invention that, when administered to a subject, is effective to (1 ) at least partially alleviating, inhibiting, preventing and/or ameliorating a condition, or a disorder or a disease (i) mediated by Btk, or (ii) associated with Btk activity, or (iii) characterized by activity (normal or abnormal) of Btk; or (2) reducing or inhibiting the activity of Btk; or (3) reducing or inhibiting the expression of Btk.
  • a therapeutically effective amount refers to the amount of the compound of the present invention that, when administered to a cell, or a tissue, or a non-cellular biological material, or a medium, is effective to at least partially reducing or inhibiting the activity of Btk; or reducing or inhibiting the expression of Btk partially or completely.
  • the term "subject" refers to an animal. Typically the animal is a mammal. A subject also refers to for example, primates (e.g., humans, male or female), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like. In certain embodiments, the subject is a primate. In yet other embodiments, the subject is a human.
  • primates e.g., humans, male or female
  • the subject is a primate.
  • the subject is a human.
  • the term “inhibit”, “inhibition” or “inhibiting” refers to the reduction or suppression of a given condition, symptom, or disorder, or disease, or a significant decrease in the baseline activity of a biological activity or process.
  • the term “treat”, “treating” or “treatment” of any disease or disorder refers in one embodiment, to ameliorating the disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms thereof).
  • “treat”, “treating” or “treatment” refers to alleviating or ameliorating at least one physical parameter including those which may not be discernible by the patient.
  • “treat”, “treating” or “treatment” refers in one embodiment, to ameliorating the disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms thereof).
  • “treat”, “treating” or “treatment” refers to alleviating or ameliorating at least one physical parameter including those which may not be discernible by the patient.
  • treatment refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both.
  • “treat”, “treating” or “treatment” refers to preventing or delaying the onset or development or progression of the disease or disorder.
  • a subject is "in need of” a treatment if such subject would benefit biologically, medically or in quality of life from such treatment.
  • any asymmetric atom (e.g. , carbon or the like) of the compound(s) of the present invention can be present in racemic or enantiomerically enriched, for example the (R)-, (S)- or (R,S)- configuration.
  • each asymmetric atom has at least 50 % enantiomeric excess, at least 60 % enantiomeric excess, at least 70 %
  • a compound of the present invention can be in the form of one of the possible isomers, rotamers, atropisomers, tautomers or mixtures thereof, for example, as substantially pure geometric (c/ ' s or trans) isomers, diastereomers, optical isomers (antipodes), racemates or mixtures thereof.
  • Any resulting mixtures of isomers can be separated on the basis of the physicochemical differences of the constituents, into the pure or substantially pure geometric or optical isomers, diastereomers, racemates, for example, by chromatography and/or fractional crystallization.
  • Any resulting racemates of final products or intermediates can be resolved into the optical antipodes by known methods, e.g., by separation of the diastereomeric salts thereof, obtained with an optically active acid or base, and liberating the optically active acidic or basic compound.
  • a basic moiety may thus be employed to resolve the compounds of the present invention into their optical antipodes, e.g., by fractional crystallization of a salt formed with an optically active acid, e.g., tartaric acid, dibenzoyl tartaric acid, diacetyl tartaric acid, di-0,0'-p-toluoyl tartaric acid, mandelic acid, malic acid or camphor-10-sulfonic acid. Racemic products can also be resolved by chiral chromatography, e.g., high pressure liquid chromatography (HPLC) using a chiral adsorbent.
  • HPLC high pressure liquid chromatography
  • the compounds of the present invention can also be obtained in the form of their hydrates, or include other solvents used for their crystallization.
  • the compounds of the present invention may inherently or by design form solvates with pharmaceutically acceptable solvents (including water); therefore, it is intended that the invention embrace both solvated and unsolvated forms.
  • solvate refers to a molecular complex of a compound of the present invention
  • solvent molecules are those commonly used in the pharmaceutical art, which are known to be innocuous to the recipient, e.g., water, ethanol, and the like.
  • solvent molecules are those commonly used in the pharmaceutical art, which are known to be innocuous to the recipient, e.g., water, ethanol, and the like.
  • hydrate refers to the complex where the solvent molecule is water.
  • the compounds of the present invention including salts, hydrates and solvates thereof, may inherently or by design form polymorphs.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of the present invention and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition can be formulated for particular routes of administration such as oral administration, parenteral administration, and rectal administration, etc.
  • the pharmaceutical compositions of the present invention can be made up in a solid form (including without limitation capsules, tablets, pills, granules, powders or suppositories), or in a liquid form (including without limitation solutions, suspensions or emulsions).
  • compositions can be subjected to conventional pharmaceutical operations such as sterilization and/or can contain conventional inert diluents, lubricating agents, or buffering agents, as well as adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers and buffers, etc.
  • the pharmaceutical compositions are tablets or gelatin capsules comprising the active ingredient together with a) diluents, e.g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine; b) lubricants, e.g., silica, talcum, stearic acid, its magnesium or calcium salt and/or polyethyleneglycol; for tablets also c) binders, e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone; if desired d) disintegrants, e.g., starches, agar, alginic acid or its sodium salt, or effervescent mixtures; and/or e) absorbents, colorants, flavors and sweeteners.
  • diluents e.g., lactose, dextrose, sucrose
  • Tablets may be either film coated or enteric coated according to methods known in the art.
  • compositions for oral administration include an effective amount of a compound of the invention in the form of tablets, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs.
  • Compositions intended for oral use are prepared according to any method known in the art for the manufacture of pharmaceutical compositions and such compositions can contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets may contain the active ingredient in admixture with nontoxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients are, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example, starch, gelatin or acacia; and lubricating agents, for example magnesium stearate, stearic acid or talc.
  • the tablets are uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate can be employed.
  • Formulations for oral use can be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example, peanut oil, liquid paraffin or olive oil.
  • compositions are aqueous isotonic solutions or suspensions, and suppositories are advantageously prepared from fatty emulsions or suspensions.
  • Said compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers. In addition, they may also contain other therapeutically valuable substances.
  • Said compositions are prepared according to conventional mixing, granulating or coating methods, respectively, and contain about 0.1-75%, or contain about 1-50%, of the active ingredient.
  • compositions for transdermal application include an effective amount of a compound of the invention with a suitable carrier.
  • Carriers suitable for transdermal delivery include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host.
  • transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound of the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
  • compositions for topical application include aqueous solutions, suspensions, ointments, creams, gels or sprayable formulations, e.g., for delivery by aerosol or the like.
  • topical delivery systems will in particular be appropriate for dermal application, e.g., for the treatment of skin cancer, e.g., for prophylactic use in sun creams, lotions, sprays and the like. They are thus particularly suited for use in topical, including cosmetic, formulations well-known in the art.
  • Such may contain solubilizers, stabilizers, tonicity enhancing agents, buffers and
  • a topical application may also pertain to an inhalation or to an intranasal application. They may be conveniently delivered in the form of a dry powder (either alone, as a mixture, for example a dry blend with lactose, or a mixed component particle, for example with phospholipids) from a dry powder inhaler or an aerosol spray presentation from a pressurised container, pump, spray, atomizer or nebuliser, with or without the use of a suitable propellant.
  • a dry powder either alone, as a mixture, for example a dry blend with lactose, or a mixed component particle, for example with phospholipids
  • the present invention further provides anhydrous pharmaceutical compositions and dosage forms comprising the compounds of the present invention as active ingredients, since water may facilitate the degradation of certain compounds.
  • Anhydrous pharmaceutical compositions and dosage forms of the invention can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
  • An anhydrous pharmaceutical composition may be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions are packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e. g., vials), blister packs, and strip packs.
  • compositions and dosage forms that comprise one or more agents that reduce the rate by which the compound of the present invention as an active ingredient will decompose.
  • agents which are referred to herein as “stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers, etc.
  • the compounds of formula I in free form or in salt form exhibit valuable pharmacological properties, e.g. Btk modulating properties, e.g. as indicated by in vitro and in vivo tests as provided in the next sections and are therefore indicated for therapy.
  • Btk modulating properties e.g. as indicated by in vitro and in vivo tests as provided in the next sections and are therefore indicated for therapy.
  • Compounds of the invention may be useful in the treatment of an indication selected from: Autoimmune disorders, inflammatory diseases, allergic diseases, airway diseases, such as asthma and chronic obstructive pulmonary disease (COPD), transplant rejection; diseases in which antibody production, antigen presentation, cytokine production or lymphoid organogenesis are abnormal or are undesirable; including rheumatoid arthritis, systemic onset juvenile idiopathic arthritis (SOJIA), gout, pemphigus vulgaris, idiopathic thrombocytopenic purpura, systemic lupus
  • erythematosus erythematosus
  • multiple sclerosis myasthenia gravis
  • Sjogren's syndrome autoimmune hemolytic anemia
  • vasculitides cryoglobulinemia, thrombotic thrombocytopenic purpura, chronic autoimmune urticaria, allergy (atopic dermatitis, contact dermatitis, allergic rhinitis), atherosclerosis, type 1 diabetes, type 2 diabetes, inflammatory bowel disease, ulcerative colitis, morbus Crohn, pancreatitis, glomerolunephritis, Goodpasture's syndrome, Hashimoto's thyroiditis, Grave's disease, antibody-mediated transplant rejection (AMR), graft versus host disease, B cell-mediated hyperacute, acute and chronic transplant rejection; thromboembolic disorders, myocardial infarct, angina pectoris, stroke, ischemic disorders, pulmonary embolism; cancers of haematopoietic origin including but not limited to multiple myeloma; leukemia; acute myelogenous leukemia; chronic myelogenous leukemia; lymphocytic leukemia; myeloid leukemia
  • the present invention provides the use of a compound of formula (I) or a salt thereof in therapy.
  • the therapy is selected from a disease which may be treated by inhibition of Btk.
  • the disease is selected from the afore-mentioned list, suitably rheumatoid arthritis, systemic onset juvenile idiopathic arthritis (SOJIA), pemphigus vulgaris, idiopathic
  • thrombocytopenic purpura systemic lupus erythematosus, multiple sclerosis, myasthenia gravis, Sjogren's syndrome, autoimmune hemolytic anemia, anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitides, cryoglobulinemia, thrombotic thrombocytopenic purpura, chronic autoimmune urticaria, atopic dermatitis, allergic rhinitis, type 1 diabetes, type 2 diabetes, inflammatory bowel disease, morbus Crohn, Goodpasture's syndrome, Grave's disease, antibody-mediated transplant rejection (AMR), B cell-mediated hyperacute, acute and chronic transplant rejection; multiple myeloma; acute myelogenous leukemia; chronic myelogenous leukemia; lymphocytic leukemia; myeloid leukemia; non-Hodgkin lymphoma; myelofibrosis with myeloid metaplasia; and Waldenstroem disease, more suitably
  • the invention provides a method of treating a disease which is treated by inhibition of Btk kinase comprising administration of a therapeutically acceptable amount of a compound of formula (I) or a salt thereof.
  • the disease is selected from the afore-mentioned list, suitably rheumatoid arthritis, systemic onset juvenile idiopathic arthritis (SOJIA), pemphigus vulgaris, idiopathic thrombocytopenic purpura, systemic lupus erythematosus, multiple sclerosis, myasthenia gravis, Sjogren's syndrome, autoimmune hemolytic anemia, anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitides, cryoglobulinemia, thrombotic thrombocytopenic purpura, chronic autoimmune urticaria, atopic dermatitis, allergic rhinitis, type 1 diabetes, type 2 diabetes, inflammatory bowel disease, morbus Crohn,
  • SOJIA system
  • Agents of the invention may be prepared by a reaction sequence (shown below) involving Suzuki coupling of a protected boronic esters II with the corresponding aryl halides ⁇ , conveniently furnishing intermediate III.
  • a reaction sequence shown below
  • III e.g. with diluted hydrochloric acid in methanol or the like
  • acylation e.g. with an appropriate acetylating agent, e.g.
  • the compounds of formula (I) may also be prepared by a reaction sequence involving Suzuki coupling of boronic esters IX with the corresponding aryl halides ⁇ , followed by an additional Suzuki coupling of V with boronic esters VI and an acylation of VII (optionally followed by a deprotection step), as shown in Reaction Scheme 2 below:
  • the compounds of formula (I) may also be prepared by a reaction sequence involving Suzuki coupling of boronic esters VI with the corresponding aryl halides III, acylation of intermediate X, followed by deprotection of XI and acylation of XII (optionally followed by a deprotection step), as shown in Reaction Scheme 3 below, wherein X denotes N, and the group PG refers to a protecting group that may be easily removed, such as for example, tert-butyloxycarbonyl.
  • the compounds of formula (I) may also be prepared by a reaction sequence involving Suzuki coupling of boronic esters XIII with the corresponding aryl halides III, followed by deprotection of XI and acylation of XII (optionally followed by a deprotection step), as shown in Reaction Scheme 4 below, wherein X denotes N, and the group PG refers to a protecting group that may be easily removed, such as for example, tert-butyloxycarbonyl.
  • Reaction Scheme 4 Reaction Scheme 4:
  • the compounds of formula (I) may also be prepared by a Suzuki coupling of boronic esters XIV with the corresponding aryl halides III (optionally followed by a deprotection step), as shown in Reaction Scheme 5 below:
  • the compounds of formula (I) may also be prepared by urea formation reaction of anilines VII (optionally followed by a deprotection step), as shown in Reaction Scheme 6 below:
  • the compounds of formula (I) may be prepared by a reaction sequence involving Suzuki coupling of boronic esters XIII with the corresponding aryl halides XVI. Protection of XVII is followed by halogenation XVII, deprotection of XIX, and Suzuki coupling of halide XX with boronic ester IX (optionally followed by a deprotection step), as shown in Reaction Scheme 7 below, wherein the group PG refers to a protecting group that may be easily removed, such as for example, benzenesulfonyl.
  • COMU (1-Cyano-2-ethoxy-2-oxoethylidenaminooxy)dimethylamino-morpholino- carbenium hexafluorophosphate
  • DIPEA Ethyl-diisopropyl-amine, Hunig's base, DIEA
  • HATU 0-(7-Azabenzotriazol- 1 -yl)-N , N , N ' , N '- tetramethyluroniumhexafluorophosphat
  • TFA Trifluoro-acetic acid
  • THF Tetrahydrofuran
  • Solvent A Water containing 1 mM ammonium hydrogen carbonate
  • Solvent B Acetonitrile containing 0.04% formic acid.
  • Solvent A Water containing 0.05% ammonium acetate and 0.05% formic acid.
  • Solvent B Acetonitrile containing 0.04% formic acid.
  • Example 16 was prepared analogue to Example 37 step 8 by replacing Intermediate 31 with Intermediate 9.
  • Example 17 was prepared analogue to Example 16 by replacing Intermediate 8 in step 3 with 4-cyclopropylbenzoic acid.
  • Example 18 was prepared analogue to Intermediate 9 by replacing Intermediate 7 with Intermediate 6 and Intermediate 8 with 4-(2,2,2-trifluoro-1-hydroxy-1-methyl-ethyl)- benzoic acid (WO2007/145834).
  • Example 19 was prepared analogue to Example 18 by replacing 4-(2,2,2-trifluoro-1- hydroxy-1-methyl-ethyl)-benzoic acid with 4-acetyl-benzoic acid.
  • Example 21 was prepared analogue to Intermediate 9 by replacing Intermediate 7 with Intermediate 10 and Intermediate 8 with 4-(2-hydroxy-1 ,1-dimethyl-ethyl)-benzoic acid.
  • Intermediate 11 was prepared analogue to Intermediate 1 by replacing 4-(4, 4,5,5- tetramethyl-[1 ,2,3]dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester with 3,6-dihydro-2H-pyran-4-boronic acid pinacolester.
  • Example 22 was prepared analogue to Intermediate 9 by replacing Intermediate 7 with Intermediate 12.
  • Example 23 was prepared analogue to Example 16 step 4 by replacing Intermediate 9 with Intermediate 14.
  • Example 24 was prepared analogue to Example 15 by replacing Intermediate 6 with Intermediate 16 and 4-dimethyaminobenzoyl chloride with tert-butylbenzoyl chloride.
  • Example 24 (160 mg, 0.32 mmol) was dissolved in acetic acid (5 ml) and hydrogen peroxide (0.033 ml, 0.32 mmol) was added. The mixture was stirred at room temperature for 2 hours. The mixture was treated with sodium hydrogen sulfite solution (10%, 10 ml) for 10 minutes, diluted with water, basified with 2N sodium hydroxide solution and extracted with EtOAc. The organic layer was washed with brine (2x), dried over sodium sulfate, filtered and evaporated.
  • Example 24 (160 mg, 0.32 mmol) was dissolved in DCM (15 ml), then trifluoro acetic acid (5 ml) and hydrogen peroxide (0.065 ml, 0.64 mmol) were added. The mixture was stirred at room temperature for 2 hours. The mixture was treated with sodium hydrogen sulfite solution (10%, 10 ml) for 10 minutes, diluted with water, basified with 2N sodium hydroxide solution and extracted with EtOAc. The organic layer was washed with brine (2x), dried over sodium sulfate, filtered and evaporated.
  • Intermediate 17 was prepared analogue to Intermediate 1 by replacing 4-(4, 4,5,5- tetramethyl-[1 ,2,3]dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester with 1 ,4-dioxaspiro[5,5]dec-7-en-8-boronic acid pinacol ester.
  • Example 27 was prepared analogue to Example 15 by replacing Intermediate 6 with Intermediate 18 and 4-dimethyaminobenzoyl chloride with 4-tert-butylbenzoyl chloride.
  • Example 27 A mixture of Example 27 (450 mg, 0.832 mmol) and TFA (6 ml) in DCM (30ml) was stirred at r.t. for 6 hrs. The solvents were removed in vacuo and the crude product was purified by flash chromatography (silica gel, EtOAc/MeOH/NH 4 OH gradient) to yield Intermediate 19.
  • Example 34 was prepared analogue to Example 30 by replacing Intermediate 22 with Intermediate 24.
  • Example 35 was prepared analogue to Example 31 by replacing Intermediate 22 with Intermediate 24.
  • Example 36 was prepared analogue to Example 32 by replacing Example 31 with Example 35.
  • Example 38 was prepared analogue to Example 37 step 8 by replacing Intermediate 31 with Intermediate 32.
  • Example 39 was prepared analogue to Example 37 by replacing the boronic ester Intermediate 28 in step 5 with the boronic ester Intermediate 36.
  • Example 40 was prepared analogue to Intermediate 30 in Example 37 step 6 by replacing Intermediate 28 in Example 37 step 5 with Intermediate 38.
  • Example 41 was prepared analogue to Example 37 step 7 by replacing Intermediate 30 with Example 40.
  • Example 40 To a solution of Example 40 (90 mg, 0.186 mmol) and DIPEA (0.098 ml, 0.558 mmol) in THF (10 ml) was added methanesulfonyl chloride (0.015 ml, 0.186 mmol) dropwise. The resulting mixture was stirred at r.t. for 1 hr, then quenched with water and diluted with EtOAc. The organic layer was washed with sat. aqueous NaHC0 3 solution and brine, dried with Na 2 S0 4 , and filtered. The solvents were removed in vacuo, and the crude product was purified by reversed phase HPLC (MeCN/H 2 0 gradient) to yield Example 42 as a light yellow solid.
  • Methanesulfonyl chloride 0.015 ml, 0.186 mmol
  • Example 43 was prepared analogue to Example 42 by replacing methanesulfonyl chloride with ⁇ , ⁇ -dimethylamidosulfamoyl chloride.
  • Example 45 was prepared analogue to Intermediate 6 by replacing Intermediate 5 with Intermediate 40.
  • Example 46 was prepared analogue to Intermediate 6 by replacing Intermediate 5 with Intermediate 41.
  • Example 47 was prepared analogue to Intermediate 6 by replacing Intermediate 5 with Intermediate 45, followed by basic removal of the acetate protecting group with LiOH in MeOH/water.
  • Example 48 was prepared analogue to Intermediate 29 by replacing Intermediate 1 with Intermediate 11 , followed by removal of the TBDPS protecting group with TBAF in THF.
  • Example 49 was prepared analogue to Intermediate 6 by replacing Intermediate 3 with Intermediate 11 and Intermediate 5 with Intermediate 38.
  • Example 50 was prepared analogue to Example 49 by replacing Intermediate 38 with Intermediate 47.
  • Example 51 For the Suzuki coupling between chloride Intermediate 15 (0.25 g, 0.99 mmol) and the boronic ester Intermediate 48 (0.78 g, 1.98 mmol) the same protocol was used as described in Example 1 step 1 to afford Example 51 as a beige solide.
  • Example 51 Compound of Example 51 (250 mg, 0.51 mmol) was dissolved in DCM (5 ml), then trifluoro acetic acid (5 ml) and hydrogen peroxide (0.079 ml, 0.78 mmol) were added. The mixture was stirred at room temperature for 2 hours. The mixture was treated with sodium hydrogen sulfite solution 10 % (10 ml) for 10 minutes, diluted with water, basified with 2N sodium hydroxide solution and extracted with EtOAc. The organic layer was washed with brine (2x), dried over sodium sulfate, filtered and evaporated. The residue was purified by flash chromatography on silica (EtOAc to EtOAc/MeOH/NH 4 OH 98:2:0.2) to afford the compound Example 53 as a beige solid.
  • Example 54 was prepared analogue to Intermediate 29 by replacing Intermediate 1 with Intermediate 17, followed by removal of the TBDPS protecting group with TBAF in THF.
  • Example 57 was purified by flash chromatography (siliga gel, EtOAc/MeOH gradient) to afford Example 57 as beige solid.
  • Example 58 was prepared analogue to Example 57 by replacing 3-tert-butoxy-azetidine hydrochloride with 3-isopropoxy-azetidine hydrochloride.
  • Example 59 was prepared analogue to Example 57 by replacing 3-tert-butoxy-azetidine hydrochloride with 5-fluoro-2,3-dihydro-1 H-isoindole.
  • Example 60 was prepared analogue to Example 57 by replacing 3-tert-butoxy-azetidine hydrochloride with 3-(2,2,2-trifluoro-1-trifluoromethyl-ethoxy)-azetidine hydrochloride (WO2009/077334).
  • Example 61 was prepared analogue to Example 57 by replacing Intermediate 6 with Intermediate 12.
  • Example 62 was prepared analogue to Example 57 by replacing Intermediate 6 with Intermediate 10.
  • Intermediate 10 was prepared analogue to Intermediate 6 by replacing Intermediate 5 with 5-fluoro-2-methyl-3-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)aniline.
  • Example 62 was prepared analogue to Example 57 by replacing Intermediate 6 with Intermediate 50.
  • Example 64 was prepared analogue to Intermediate 11 by replacing 4-chloro-6-iodo- 7H-pyrrolo[2,3-d]pyrimidine with Intermediate 54.
  • Example 65 was prepared analogue to Intermediate 17 by replacing 4-chloro-6-iodo- 7H-pyrrolo[2,3-d]pyrimidine with Intermediate 54.
  • Example 66 was prepared analogue to Example 28 step 2 by replacing Intermediate 19 in with Intermediate 55.
  • Example 67 was prepared analogue to Example 56 by replacing Intermediate 49 with Intermediate 55.
  • the inhibitory activity of the present compounds against Btk was assessed in a biochemical enzyme assay.
  • Assay plates in 384 well format were prepared with 8-point serial dilutions for the test compounds on a Thermo CatX workstation equipped with a Innovadyne Nanodrop Express.
  • the assay plates were prepared by addition of 50 nl per well of compound solution in 90 % DMSO.
  • kinase reactions were started by stepwise addition of 4.5 ⁇ per well of peptide/ATP-solution (4 ⁇ FITC-Ahx- TSELKKVVALYDYMPMNAND-NH2, 164 ⁇ ATP) in kinase buffer (50mM HEPES, pH 7.5, 1 mM DTT, 0.02% Tween20, 0.02% BSA, 0.6% DMSO, 10 mM beta- glycerophosphate, and 10 ⁇ sodium orthovanadate, 18 mM MgCI2, 1 mM MnCI2) and 4.5 ⁇ per well of enzyme solution (6.4nM full-lenght human recombinant BTK) in kinase buffer.
  • peptide/ATP-solution 4 ⁇ FITC-Ahx- TSELKKVVALYDYMPMNAND-NH2, 164 ⁇ ATP
  • kinase buffer 50mM HEPES, pH 7.5, 1 mM DTT, 0.02% Tween
  • Kinase reactions were incubated at 30°C for 60 minutes and subsequently terminated by addition of 16 ⁇ per well of stop solution (100 mM HEPES pH 7.5, 5 % DMSO, 0.1 % Caliper coating reagent, 10 mM EDTA, and 0.015 % Brij35).
  • Stop solution 100 mM HEPES pH 7.5, 5 % DMSO, 0.1 % Caliper coating reagent, 10 mM EDTA, and 0.015 % Brij35.
  • Kinase reactions were analyzed on a Caliper LC3000 workstation by separating phosphorylated and unphosphorylated peptides and kinase activities were calculated from the amounts of newly formed phospho-peptide.
  • Inhibition data were calculated by comparison to control reactions without enzyme (100 % inhibition) and without inhibitors (0 % inhibition). The concentration of inhibitor required for 50 % inhibition (IC50) was calculated from the inhibition in response to inhibitor concentrations.
  • Example 44 0.017
  • Example 45 0.008
  • the present compounds might also be assessed for their capacity to inhibit Btk-dependent FcG receptor-induced IL-8 secretion in human cells.
  • the human myeloid leukemia THP1 cell line (ATCC TIB202) was grown in RPMI 1640 medium
  • tissue-culture grade 384-well plates was coated with human IgG of unknown specificity by incubating overnight at 4°C with 40 ⁇ /well of a 50 ⁇ g/ml IgG solution in PBS. On the day of the experiment, plates were washed 5 times with 80 ⁇ water on a Molecular Devices Aquamax DW4 plate washer. Solutions of the test compounds in 90 % DMSO were added to each well on a Hamilton Microlab Star liquid handling station to 40 ⁇ /well tissue culture medium and the total DMSO concentration was adjusted to 0.1 %.
  • Differentiated THP1 cells were then added in 40 ⁇ /well to reach a final density of 5 ⁇ 00 cells/well in 80 ⁇ culture medium. After 24 hours, IL-8 secretion was measured in the supernatant by the IL-8 HTRF assay following the protocol of the vendor (CisBio international). Inhibition data were calculated by comparison to control cultures without IgG stimulus enzyme (100 % inhibition) and without inhibitors (0 % inhibition). The concentration of inhibitor required for 50 % inhibition (IC50) was calculated from the inhibition in response to inhibitor concentrations.
  • the inhibitory activity of the present compounds in blood was assessed in the following in vitro B cell activation assay.
  • Whole blood was collected from the abdominal aorta of anaesthetized adult male Lewis rats and was anticoagulated with 100 U/ml sodium heparin. Blood was then diluted to 50 % with high glucose DMEM
  • compounds of the invention may generally be useful in the treatment of an indication selected from:
  • Autoimmune disorders inflammatory diseases, allergic diseases, airway diseases, such as asthma and chronic obstructive pulmonary disease (COPD), transplant rejection; diseases in which antibody production, antigen presentation, cytokine production or lymphoid organogenesis are abnormal or are undesirable; including rheumatoid arthritis, systemic onset juvenile idiopathic arthritis (SOJIA), gout, pemphigus vulgaris, idiopathic thrombocytopenic purpura, systemic lupus erythematosus, multiple sclerosis, myasthenia gravis, Sjogren's syndrome, autoimmune hemolytic anemia, anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitides, cryoglobulinemia, thrombotic thrombocytopenic purpura, chronic autoimmune urticaria, allergy (atopic dermatitis, contact dermatitis, allergic rhinitis), atherosclerosis, type 1 diabetes, type 2 diabetes, inflammatory bowel disease, ulcer
  • glomerolunephritis Goodpasture's syndrome, Hashimoto's thyroiditis, Grave's disease, antibody-mediated transplant rejection (AMR), graft versus host disease, B cell-mediated hyperacute, acute and chronic transplant rejection; thromboembolic disorders, myocardial infarct, angina pectoris, stroke, ischemic disorders, pulmonary embolism; cancers of haematopoietic origin including but not limited to multiple myeloma; a leukaemia; acute myelogenous leukemia; chronic myelogenous leukemia; lymphocytic leukemia; myeloid leukemia; non-Hodgkin lymphoma; lymphomas; polycythemia vera; essential thrombocythemia; myelofibrosis with myeloid metaplasia; and Waldenstroem disease.
  • AMR antibody-mediated transplant rejection
  • graft versus host disease B cell-mediated hyperacute, acute and chronic transplant rejection
  • the therapy is selected from a disease which may be treated by an antagonist of Bruton's tyrosine kinase.
  • the invention provides a method of treating a disease which is treated by the modulation of Btk- comprising administration of a therapeutically acceptable amount of a compound of formula (I) or a salt thereof.
  • the disease is selected from the afore-mentioned lists
  • the compound of the present invention may be administered either simultaneously with, or before or after, one or more other therapeutic agent.
  • the compound of the present invention may be administered separately, by the same or different route of
  • the compounds of formula (I) may be administered as the sole active ingredient or in conjunction with, e.g. as an adjuvant to, other drugs e.g. immunosuppressive or immunomodulating agents or other anti-inflammatory agents, e.g. for the treatment or prevention of alio- or xenograft acute or chronic rejection or inflammatory or autoimmune disorders, or a chemotherapeutic agent, e.g a malignant cell anti-proliferative agent.
  • the compounds of formula (I) may be used in combination with a calcineurin inhibitor, e.g. cyclosporin A or FK 506; a mTOR inhibitor, e.g.
  • rapamycin 40-O-(2- hydroxyethyl)-rapamycin, CCI779, ABT578, AP23573, AP23464, AP23675, AP23841 , TAFA-93, biolimus-7 or biolimus-9; an ascomycin having immunosuppressive properties, e.g. ABT-281 , ASM981 , etc.; corticosteroids; cyclophosphamide; azathioprene; methotrexate; leflunomide; mizoribine; mycophenolic acid or salt; mycophenolate mofetil; 15-deoxyspergualine or an immunosuppressive homologue, analogue or derivative thereof; a PKC inhibitor, e.g.
  • a JAK3 kinase inhibitor e.g. N-benzyl-3,4-dihydroxy- benzylidene-cyanoacetamide a-cyano-(3,4-dihydroxy)-]N-benzylcinnamamide
  • mono-citrate also called CP-690,550
  • sphingosine-1-phosphate receptor modulators such as FTY720 (fingolimod), or compounds disclosed in WO 2005/000833
  • immunosuppressive monoclonal antibodies e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC, CD2, CD3, CD4, CD7, CD8, CD25, CD28, CD40, CD45, CD52, CD58, CD80, CD86 or their ligands
  • other immunomodulatory compounds e.g.
  • a recombinant binding molecule having at least a portion of the extracellular domain of CTLA4 or a mutant thereof, e.g. an at least extracellular portion of CTLA4 or a mutant thereof joined to a non-CTLA4 protein sequence, e.g. CTLA4lg (for ex. designated ATCC 68629) or a mutant thereof, e.g. LEA29Y; adhesion molecule inhibitors, e.g. LFA- 1 antagonists, ICAM-1 or -3 antagonists, VCAM-4 antagonists or VLA-4 antagonists; or a chemotherapeutic agent, e.g.
  • a compound of formula (I) may be selected from a PI3K inhibitor (e.g. pan, or alpha, beta, gamma, delta selectives), TNF inhibitors, ILI beta inhibitors, IL17 inhibitors, and inhibitors of IL6 or IL receptor.
  • co-administration or “combined administration” or the like as utilized herein are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time.
  • pharmaceutical combination as used herein means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients.
  • fixed combination means that the active ingredients, e.g. a compound of formula (I) and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage.
  • non-fixed combination means that the active ingredients, e.g.
  • a compound of formula (I) and a co-agent are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the 2 compounds in the body of the patient.
  • cocktail therapy e.g. the administration of 3 or more active ingredients.
  • the invention provides a product comprising a compound of formula (I) and at least one other therapeutic agent as a combined preparation for simultaneous, separate or sequential use in therapy.
  • the therapy is the treatment of a disease or condition mediated by Btk kinases.
  • Products provided as a combined preparation include a composition comprising the compound of formula (I) and the other therapeutic agent(s) together in the same pharmaceutical composition, or the compound of formula (I) and the other therapeutic agent(s) in separate form, e.g. in the form of a kit.
  • the invention provides a pharmaceutical composition comprising a compound of formula (I) and another therapeutic agent(s).
  • a pharmaceutical composition comprising a compound of formula (I) and another therapeutic agent(s).
  • composition may comprise a pharmaceutically acceptable excipient, as described above.
  • the invention provides a kit comprising two or more separate pharmaceutical compositions, at least one of which contains a compound of formula (I).
  • the kit comprises means for separately retaining said compositions, such as a container, divided bottle, or divided foil packet.
  • a container, divided bottle, or divided foil packet An example of such a kit is a blister pack, as typically used for the packaging of tablets, capsules and the like.
  • the kit of the invention may be used for administering different dosage forms, for example, oral and parenteral, for administering the separate compositions at different dosage intervals, or for titrating the separate compositions against one another.
  • the kit of the invention typically comprises directions for
  • the compound of the invention and the other therapeutic agent may be manufactured and/or formulated by the same or different manufacturers. Moreover, the compound of the invention and the other therapeutic may be brought together into a combination therapy: (i) prior to release of the combination product to physicians (e.g. in the case of a kit comprising the compound of the invention and the other therapeutic agent); (ii) by the physician themselves (or under the guidance of the physician) shortly before administration; (iii) in the patient themselves, e.g. during sequential administration of the compound of the invention and the other therapeutic agent.
  • the invention provides the use of a compound of formula (I) for treating a disease or condition mediated by Btk kinases, wherein the medicament is prepared for administration with another therapeutic agent.
  • the invention also provides the use of another therapeutic agent for treating a disease or condition mediated by Btk , wherein the medicament is administered with a compound of formula (I).
  • the invention also provides a compound of formula (I) for use in a method of treating a disease or condition mediated by Btk, wherein the compound of formula (I) is prepared for administration with another therapeutic agent.
  • the invention also provides another therapeutic agent for use in a method of treating a disease or condition mediated by Btk , wherein the other therapeutic agent is prepared for administration with a compound of formula (I).
  • the invention also provides a compound of formula (I) for use in a method of treating a disease or condition mediated by Btk , wherein the compound of formula (I) is administered with another therapeutic agent.
  • the invention also provides another therapeutic agent for use in a method of treating a disease or condition mediated by Btk, wherein the other therapeutic agent is administered with a compound of formula (I).

Abstract

The present invention describes new pyrrolo pyrimidine derivatives and pharmaceutically acceptable salts thereof which appear to interact with Bruton's tyrosine kinase (Btk). Accordingly, the novel pyrrolo pyrimidines may be effective in the treatment of autoimmune disorders, inflammatory diseases, allergic diseases, airway diseases, such as asthma and chronic obstructive pulmonary disease (COPD), transplant rejection, cancers e.g. of hematopoietic origin or solid tumors.

Description

Novel Pyrrolo Pyrimidine Derivatives
The present invention describes new pyrrolo pyrimidine derivatives that are good drug candidates.
The compounds of the present invention may generally exhibit a selective inhibition of Bruton's tyrosine kinase (Btk).
Background of the invention
The essential role of Btk in autoimmune disease is underlined by the observations that Btk-deficient mice are protected in standard preclinical models for rheumatoid arthritis (Jansson and Holmdahl, 1993), systemic lupus erythematosus (Steinberg, B.J. et al., J. Clin. Invest, 70, 587-597, 1982), as well as allergic disease and anaphylaxis (Hata.D. et al., J. Exp. Med. 187, 1235-1247, 1998). In addition, many cancers and lymphomas express Btk and appear to be dependent on Btk function (Davis, R.E. et al., Nature, 463, 88-92, 2010).
Therefore, inhibition of Btk activity may be useful in the treatment of immune disorders such as rheumatoid arthritis, systemic lupus erythematosus, allergic diseases, anaphylaxis and inflammatory conditions. Moreover, inhibition of Btk may be useful in the treatment of cancers of haematopoietic origin including chronic myelogenous leukemia, myeloid leukemia, non-Hodgkin lymphoma and other B cell lymphomas.
The compounds of the present invention may therefore potentially be useful in the treatment of a wide range of disorders, particularly Btk-related diseases or disorders, and may for example be useful in the treatment of autoimmune disorders, inflammatory diseases, allergic diseases, airway diseases, such as asthma and chronic obstructive pulmonary disease (COPD), transplant rejection, or cancers e.g. of hematopoietic origin or solid tumors. More particularly, the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof;
Figure imgf000003_0001
wherein,
R1 is hydrogen, CrC6 alkyi optionally substituted by hydroxy;
R2 is hydrogen or halogen;
R3 is hydrogen or halogen;
R4 is hydrogen,
R5 is phenyl optionally substituted by halogen; SF5; NR6R7; hydroxy; C C6 alkoxy; d- C6 alkenyl; CrC6 alkyi carbonyl; CrC6 alkyi optionally substituted by hydroxy, halogen, or Ci-C6 alkoxy; or C3-C6 cycloalkyi optionally substituted by halogen, hydroxy, or C C6 alkyi optionally substituted by halogen; or
R5 is a 4 - 14 membered mono- or bicyclic heterocyclyl or heteroaryl ring system comprising 1 , 2 or 3 heteroatoms selected from N, S and O that ring being optionally substituted by halogen; hydroxy; CrC6 alkoxy optionally substituted by hydroxy or halogen; or CrC6 alkyi optionally substituted by hydroxy or halogen;
or R4 and R5 together with the atoms to which they are bound form a piperidone ring, optionally comprising an annulated phenyl ring, any such ring being optionally substituted by C C6 alkyi, C C6 alkoxy, or C3-C6 cycloalkyi each of which substitution member may optionally be substituted by halogen or hydroxy;
R6 and R7 are independently selected from hydrogen or CrC6 alkyi;
or R6 and R7 together with the nitrogen atom to which they are bound form a 4 - 8 membered saturated azacycloalkane ring, optionally substituted by halogen, hydroxy or
Ci-C6 alkyi;
X is O, S(0)n wherein n is 0, 1 or 2, or
Figure imgf000003_0002
wherein q is 2 or 3, and R10 is absent; or X is CH or N; and R10 is hydrogen, hydroxy, -NR6R7, -CO-R1 1 , -S(0)p-R12 wherein p is 1 or 2,
R 1 1 is Ci-C6 alkyl optionally substituted by hydroxy, cyano, halogen, carboxy or CrC6 alkoxy carbonyloxy; or NR6R7; and
R12 is Ci-C6 alkyl or NR6R7.
In another embodiment the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 is hydrogen, R5 is phenyl substituted by halogen; CrC6 alkoxy or CrC6 alkyl optionally substituted by halogen or hydroxy; or C3-C6 cycloalkyi optionally substituted by halogen, hydroxy, C C6 substituted by halogen; X is O, S(0)n wherein n is 0, 1 or 2, or
Figure imgf000004_0001
wherein q is 2 or 3, and R10 is absent;
and the remaining variables are as defined above.
In another embodiment the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one optionally substituted by C3-C6 cycloalkyi or C C6 substituted by hydroxy ; X is O, S(0)n wherein n is 0, 1 or 2, or
Figure imgf000004_0002
wherein q is 2 or 3, and R10 is absent; and the remaining variables are as defined above.
In another embodiment the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 is hydrogen, R5 is phenyl substituted by halogen; CrC6 alkoxy; CrC6 alkyl optionally substituted by halogen or hydroxy; or C3-C6 cycloalkyi optionally substituted by halogen, hydroxy, or d- C6 alkyl optionally substituted by halogen; X stands for O and R10 is absent; or X stands for N, and R10 is hydrogen or -CO-R1 1 , and the remaining variables are as defined above. ln another embodiment the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 is hydrogen; R5 is phenyl substituted by CrC6 alkoxy; CrC6 alkyl optionally substituted by halogen or hydroxy; or C3-C6 cycloalkyl optionally substituted by halogen, hydroxy, or CrC6 alkyl optionally substituted by halogen; X stands for N, R10 is hydrogen or -CO-R1 1 , R1 1 stands for NR6R7 wherein R6 and R7 are independently hydrogen or methyl; and the remaining variables are as defined above.
In another embodiment the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 is hydrogen, R5 is azetidine optionally substituted by CrC6 alkoxy, X stands for N, and R10 is hydrogen or -CO-R1 1 , and the remaining variables are as defined above.
In another embodiment the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one optionally substituted by C3-C6 cycloalkyl or C C6 alkyl optionally substituted by hydroxy, X stands for N, and R10 is hydrogen or -CO-R1 1 , and the remaining variables are as defined above.
In another embodiment the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one substituted by C3-C6 cycloalkyl or C C6 alkyl optionally substituted by hydroxy in the 6-position of said isoquinolin-ring, X stands for N, and R10 is hydrogen or -CO-R1 1 , and the remaining variables are as defined above.
In another embodiment the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 is hydrogen, R5 is azetidine optionally substituted by CrC6 alkoxy, X stands for O, and the remaining variables are as defined above.
In another embodiment the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one optionally substituted by C3-C6 cycloalkyi or C C6 alkyl optionally substituted by hydroxy, X stands for O, and the remaining variables are as defined above.
In another embodiment the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one substituted by C3-C6 cycloalkyi or C C6 alkyl optionally substituted by hydroxy in the 6-position of said isoquinolin-ring, X stands for O, and the remaining variables are as defined above.
In another embodiment the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is hydrogen, methyl or
hydroxymethyl , R2 and R3 are independently from each other selected from hydrogen and halogen, R4 is hydrogen, R5 is phenyl substituted one or more times by halogen, C3-C6 cycloalkyi, or CrC6 alkyl optionally substituted by hydroxy, X stands for O or S, and R10 is absent.
In another embodiment the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof wherein R1 is methyl or hydroxymethyl , R2 and R3 are independently from each other selected from hydrogen and halogen, R4 is hydrogen, R5 is phenyl substituted one or more times by halogen, C3-C6 cycloalkyi, or Ci-C6 alkyl optionally substituted by hydroxy, X stands for N, R10 is hydrogen or -CO-R1 1 , and R1 1 is NR6R7 wherein R6 and R7 are independently selected from C Ce-alkyl. With regard to a compound of formula (I) the following significances represent further embodiments of the invention independently, collectively or in any combination or in any sub-combination thereof:
1. R1 is hydrogen, methyl or hydroxymethyl;
2. R1 is methyl or hydroxymethyl;
3. R2 and R3 are independently hydrogen or fluoro;
4. R1 is methyl or hydroxymethyl and R2 and R3 are independently hydrogen or fluoro;
5. R4 is hydrogen;
6. R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one optionally substituted by C3-C6 cycloalkyi or CrC6 alkyl optionally substituted by hydroxy;
7. R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one optionally substituted by C3-C6 cycloalkyi or CrC6 alkyl optionally substituted by hydroxy in the 6- position of the 3,4-dihydro-2H-isoquinolin-1-one ring;
8. R5 is phenyl optionally substituted by -NR6R7, halogen; CrC6 alkoxy, CrC6 alkenyl, C3-C6 cycloalkyi, or CrC6 alkyl optionally substituted by halogen or hydroxy;
9. R5 is phenyl substituted by -NR6R7, halogen, CrC6 alkoxy or C C6 alkyl optionally substituted by halogen or hydroxy;
10. R5 is phenyl substituted by halogen, CrC6 alkoxy, C3-C6 cycloalkyi or C C6 alkyl optionally substituted by fluoro or hydroxy;
11. R5 is a 4-, 5-, 6-, or 7-membered monocyclic heterocycle, or a 7-, 8-, 9-, 10-, 1 1-, or 12-membered bicyclic heterocycle comprising 1 , 2 or 3 heteroatoms selected from N, S and O that ring being optionally substituted by halogen; hydroxy; CrC6 alkoxy optionally substituted by hydroxy or halogen; or C C6 alkyl optionally substituted by hydroxy or halogen;
12. R5 is a 4-, 5-, 6-, or 7-membered monocyclic heterocycle comprising 1 or 2 heteroatoms selected from N, S and O that ring being optionally substituted by halogen; hydroxy; CrC6 alkoxy optionally substituted by hydroxy or halogen; or Ci-C6 alkyl optionally substituted by hydroxy or halogen;
13. R5 is azetidine substituted by C C6 alkoxy or C C6 alkyl; 14. X is O and R10 is absent or is N and R10 is H or CO-R1 1 ;
15. X is O and R10 is absent or is N and R10 is CO-R1 1 ;
16. X is O and R10 is absent;
17. X is N and R10 is CO-R11 ;
18. R11 is NR6R7 and R6 and R7 are independently selected from hydrogen or d- C6 alkyl;
19. R11 is NR6R7 and R6 and R7 are independently selected from d-C6 alkyl;
20. R11 is NR6R7 and R6 and R7 are independently selected from C C3 alkyl;
21. R11 is NR6R7 and R6 and R7 are methyl.
In another embodiment the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof for use as a medicament.
In another embodiment the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of a disease or disorder mediated by Btk.
In another embodiment the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt thereof, which is selected from:
4-(4-{5-Fluoro-3-[4-(1-fluoro-cyclopropyl)-benzoylamino]-2-methyl-phenyl}-7H- pyrrolo[2'3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 4-(4-{3-[(3,3-Dimethyl-2,3-dihydro-benzofuran-6-carbonyl)-amino]-5-fluoro-2-methyl- phenyl}-7H-pyrrolo[2'3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-(4-{5-Fluoro-2-methyl-3-[(5-methyl-4,5,6,7-tetrahydro-benzo[b]thiophene-2-carbonyl)- amino]-phenyl}-7H-pyrrolo[2'3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-(4-{5-Fluoro-3-[4-isopropyl-methyl-amino)-benzoylamino]-2-methyl-phenyl}-7H- pyrrolo[2'3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 3-Methyl-1 H-indole-6-carboxylic acid {3-[6-(1-dimethylcarbamoyl-1 ,2,3,6-tetrahydro- pyridin-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5-fluoro-2-methyl-phenyl}-amide, 4-(4-{5-Fluoro-3-[4-(2-hydroxy-1 ,1-dimethyl-ethyl)-benzoylamino]-2-methyl-phenyl}-7H- pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 4-{4-[5-Fluoro-2-methyl-3-(4-piperidin-1-yl-benzoylamino)-phenyl]-7H-pyrrolo[2,3- d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-{4-[5-Fluoro-3-(isopropenyl-benzoylamino)-2-methyl-phenyl]-7H-pyrrolo[2,3- d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-(4-{5-Fluoro-2-methyl-3-[4-(1-trifluoromethyl-cyclopropyl)-benzoylamino]-phenyl}-7H- pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 4-{4-[5-Fluoro-3-(4-isopropoxy-benzoylamino)-2-methyl-phenyl]-7H-pyrrolo[2,3- d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-(4-{5-Fluoro-3-[4-pentafluorothio-benzoylamino]-2-methyl-phenyl}-7H-pyrrolo[2,3- d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-(4-{5-Fluoro-3-[4-(2-methoxy-1 ,1 -dimethyl-ethyl )-benzoylamino]-2-methyl-phenyl}-7H- pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 4-(4-{5-Fluoro-3-[4-(1-methoxy-1 -methyl-ethyl )-benzoylamino]-2-methyl-phenyl}-7H- pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 1-Methyl-1 H-pyrrolo[2,3-b]pyridine-6-carboxylic acid {3-[6-(1-dimethyl-carbamoyl-1 ,2,3,6- tetrahydro-pyridin-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5-fluoro-2-methyl-phenyl}-amide, 4-{4-[3-(4-Dimethylamino-benzoylamino)-5-fluoro-2-methyl-phenyl]-7H-pyrrolo[2,3- d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-(4-{3-[2-Fluoro-4-(1-hydroxy-1 -methyl-ethyl )-benzoylamino]-2-hydroxymethyl-phenyl}- 7H-pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid
dimethylamide,
4-{4-[3-(4-Cyclopropyl-benzoylamino)-2-hydroxymethyl-phenyl]-7H-pyrrolo[2,3- d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-(4-{5-Fluoro-2-methyl-3-[4-(2,2,2-trifluoro-1-hydroxy-1 -methyl-ethyl )-benzoylamino]- phenyl}-7H-pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-{4-[3-(4-Acetyl-benzoylamino)-5-fluoro-2-methyl-phenyl]-7H-pyrrolo[2,3-d]pyrimidin-6- yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-{4-[3-(4-Cyclopropyl-benzoylamino)-4-fluoro-phenyl]-7H-pyrrolo[2,3-d]pyrimidin-6-yl}- 3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-(4-{4-Fluoro-3-[4-(2-hydroxy-1 , 1-dimethyl-ethyl)-benzoylamino]-phenyl}-7H-pyrrolo[2,3- d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, N-{3-[6-(3,6-Dihydro-2H-pyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5-fluoro-2-methyl- phenyl}-2-fluoro-4-(1-hydroxy-1 -methyl-ethyl )-benzamide,
N-{3-[6-(3,6-Dihydro-2H-pyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2-hydroxymethyl- phenyl}-2-fluoro-4-(1-hydroxy-1 -methyl-ethyl )-benzamide,
4-tert-Butyl-N-{3-[6-(3,6-dihydro-2H-thiopyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5- fluoro-2-methyl-phenyl}-benzamide,
4-tert-Butyl-N-{5-fluoro-2-methyl-3-[6-(3,6-dihydro-1-oxido-2H-thiopyran-4-yl)-7H- pyrrolo[2,3-d]pyrimidin-4-yl]-phenyl}-benzamide,
4-tert-Butyl-N-{3-[6-(3,6-dihydro-1 ,1-dioxido-2H-thiopyran-4-yl)-7H-pyrrolo[2,3- d]pyrimidin-4-yl]-5-fluoro-2-methyl-phenyl}-benzamide,
4-tert-Butyl-N-{3-[6-(1 ^-dioxa-spiro[4.5]dec-7-en-8-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5- fluoro-2-methyl-phenyl}-benzamide,
4-tert-Butyl-N-{5-fluoro-3-[6-(4-hydroxy-cyclohex-1-enyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]- 2-methyl-phenyl}-benzamide,
4-{4-[3-(4-Cyclopropyl-benzoylamino)-5-fluoro-2-methyl-phenyl]-7H-pyrrolo[2'3- d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-Cyclopropyl-N-(5-fluoro-2-methyl-3-{6-[1-(pyrrolidine-1-carbonyl)-1 ,2,3,6-tetrahydro- pyridin-4-yl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}-phenyl)-benzamide,
Acetic acid 2-(4-{4-[3-(4-cyclopropyl-benzoylamino)-5-fluoro-2-methyl-phenyl]-7H- pyrrolo[2,3-d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridin-1-yl)-2-oxo-ethyl ester,
4-Cyclopropyl-N-(5-fluoro-3-{6-[1-(2-hydroxy-actyl)-1 ,2,3,6-tetrahydro-pyridin-4-yl]-7H- pyrrolo[2,3-d]pyrimidin-4-yl}-2-methyl-phenyl)benzamide,
N-(3-{6-[1-(2-Cyano-acetyl)-1 ,2,3,6-tetrahydro-pyridin-4-yl]-7H-pyrrolo[2,3-d]pyrimidin-4- yl}-5-fluoro-2-methyl-phenyl)-4-cyclopropyl-benzamide,
N-(5-Fluoro-2-methyl-3-{6-[1-(pyrrolidine-1-carbonyl)-1 ,2,3,6-tetrahydro-pyridin-4-yl]-7H- pyrrolo[2,3-d]pyrimidin-4-yl}-phenyl)-4-(pentafluoro-sulfanyl)-benzamide,
Acetic acid 2-[4-(4-{5-fluoro-2-methyl-3-[4-(pentafluoro-sulfanyl)-benzoylamino]-phenyl}-
7H-pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridin-1-yl]-2-oxo-ethyl ester,
N-(5-Fluoro-3-{6-[1-(2-hydroxy-acetyl)-1 ,2,3,6-tetrahydro-pyridin-4-yl]-7H-pyrrolo[2,3- d]pyrimidin-4-yl}-2-methyl-phenyl)-4-(pentafluoro-sulfanyl)-benzamide,
4-{4-[3-(4-tert-Butyl-benzoylamino)-2-(tert-butyl-diphenyl-silanyloxymethyl)-phenyl]-7H- pyrrolo[2,3-d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester,
4-tert-Butyl-N-(3-{6-[1-(2-fluoro-acetyl)-1 ,2,3,6-tetrahydro-pyridin-4-yl]-7H-pyrrolo[2,3- d]pyrimidin-4-yl}-2-hydroxymethyl-phenyl)-benzamide, 4-(4-{5-Fluoro-3-[2-fluoro-4-(1-hydroxy-1 -methyl-ethyl )-benzoylamino]-2-methyl-phenyl}- 7H-pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid
dimethylamide,
4-tert-Butyl-N-{5-fluoro-2-methyl-3-[6-(1 ,2,3,6-tetrahydro-pyridin-4-yl)-7H-pyrrolo[2,3- d]pyrimidin-4-yl]-phenyl}-benzamide,
4-{4-[3-(4-tert-Butyl-benzoylamino)-5-fluoro-2-methyl-phenyl]-7H-pyrrolo[2,3-d]pyrimidin- 6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-tert-Butyl-N-{5-fluoro-3-[6-(1-methanesulfonyl-1 ,2,3,6-tetrahydro-pyridin-4-yl)-7H- pyrrolo[2,3-d]pyrimidin-4-yl]-2-methyl-phenyl}-benzamide,
4-tert-Butyl-N-{3-[6-(1-dimethylsulfamoyl-1 ,2,3,6-tetrahydro-pyridin-4-yl)-7H-pyrrolo[2,3- d]pyrimidin-4-yl]-5-fluoro-2-methyl-phenyl}-benzamide,
4-tert-Butyl-N-{3-[6-(1-methanesulfonyl-1 ,2,3,6-tetrahydro-pyridin-4-yl)-7H-pyrrolo[2,3- d]pyrimidin-4-yl]-2-methyl-phenyl}-benzamide,
4-{4-[3-(6-tert-Butyl-1 -oxo-3, 4-dihydro-1 H-isoquinolin-2-yl )-2-hydroxymethyl-phenyl]-7H- pyrrolo[2,3-d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 4-{4-[3-(6-Cyclopropyl-1 -oxo-3, 4-dihydro-1 H-isoquinolin-2-yl )-2-hydroxymethyl-phenyl]- 7H-pyrrolo[2,3-d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid
dimethylamide,
4-(4-{2-Hydroxymethyl-3-[6-(1-hydroxy-1 -methyl-ethyl )-1 -oxo-3, 4-dihydro-1 H-isoquinolin- 2-yl]-phenyl}-7H-pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-tert-Butyl-N-{3-[6-(3,6-dihydro-2H-pyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2- hydroxymethyl-phenyl}-benzamide,
4-tert-Butyl-N-{3-[6-(3,6-dihydro-2H-pyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5-fluoro- 2-methyl-phenyl}-benzamide,
N-{3-[6-(3,6-Dihydro-2H-pyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5-fluoro-2-methyl- phenyl}-4-dimethylamino-benzamide,
4-tert-Butyl-N-{3-[6-(3,6-dihydro-2H-thiopyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2- methyl-phenyl}-benzamide,
4-tert-Butyl -N-{2-methyl-3-[6-(3,6-dihydro-1-oxido-2H-thiopyran-4-yl)-7H-pyrrolo[2,3- d]pyrimidin-4-yl]-phenyl}-benzamide,
4-tert-Butyl -N-{3-[6-(3,6-dihydro-1 ,1-dioxido-2H-thiopyran-4-yl)-7H-pyrrolo[2,3- d]pyrimidin-4-yl]-2-methyl-phenyl}-benzamide, 4-tert-Butyl-N-{3-[6-(1 ^-dioxa-spiro[4.5]dec-7-en-8-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2- hydroxymethyl-phenyl}-benzamide,
4 ert-Butyl-N 3-[6-(4-hydroxy-cyclohex-1-enyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2- hydroxymethyl-phenyl}-benzamide,
4 ert-Butyl-N-{3-[6-(4-dimethylamino-cyclohex-1-eny^
hydroxymethyl-phenyl}-benzamide,
4-(4 3-[(3 ert-Butoxy-azetidine-1-carbonyl)-amino]-5-fluoro-2-methyl-phenyl}-7H- pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4- (4 5-Fluoro-3-[(3-isopropoxy-azetidine-1-carbonyl)-amino]-2-methyl-phenyl}-7H- pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
5- Fluoro-1 ,3-dihydro-isoindole-2-carboxylic acid {3-[6-(1-dimethylcarbamoyl-1 ,2,3,6- tetrahydro-pyridin-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5-fluoro-2-methyl-phenyl}-amide, 4-[4-(5-Fluoro-2-methyl-3-{[3-(2,2,2-trifluoro-1-trifluoromethyl-ethoxy)-azetidine-1- carbonyl]-amino}-phenyl)-7H-pyrrolo[2,3-d]pyrimidin-6-yl]-3,6-dihydro-2H-pyridine-1- carboxylic acid dimethylamide,
3- tert-Butoxy-azetidine-1-carboxylic acid {3-[6-(3,6-dihydro-2H-pyran-4-yl)-7H- pyrrolo[2,3-d]pyrimidin-4-yl]-5-fluoro-2-methyl-phenyl}-amide,
4- (4-{3-[(3-tert-Butoxy-azetidine-1-carbonyl)-amino]-4-fluoro-phenyl}-7H-pyrrolo[2,3- d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-(4-{3-[(3-tert-Butoxy-azetidine-1-carbonyl)-amino]-4-fluoro-2-methyl-phenyl}-7H- pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 4-tert-Butyl-N-{3-[6-(3,6-dihydro-2H-pyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2- methyl-phenyl}-benzamide,
4 ert-Butyl-N-{3-[6-(1 ,4-dioxa-spiro[4.5]dec-7-en-8-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2- methyl-phenyl}-benzamide,
4-tert-Butyl-N-{3-[6-(4-hydroxy-cyclohex-1-enyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2- methyl-phenyl}-benzamide, and
4-tert-Butyl-N-{3-[6-(4-dimethylamino-cyclohex-1-enyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2- methyl-phenyl}-benzamide.
As used herein, the term "alkyl" refers to a fully saturated branched or unbranched hydrocarbon moiety having up to 20 carbon atoms. Unless otherwise provided, alkyl refers to hydrocarbon moieties having 1 to 16 carbon atoms, 1 to 10 carbon atoms, 1 to 7 carbon atoms, or 1 to 4 carbon atoms. Representative examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, /'so-propyl, n-butyl, sec-butyl, /'so-butyl, tert- butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, 3-methylhexyl, 2,2- dimethylpentyl, 2,3- dimethylpentyl, n-heptyl, n-octyl, n-nonyl, n-decyl and the like.
As used herein, the term "alkenyl" refers to an unsaturated branched or unbranched hydrocarbon moiety having 2 to 20 carbon atoms. It comprises 2 to 20 carbon atoms, Unless otherwise provided, alkenyl refers to moieties having 2 to 16 carbon atoms, 2 to 10 carbon atoms, 2 to 7 carbon atoms, or 2 to 4 carbon atoms. Representative examples of alkenyl include, but are not limited to, ethenyl, n-propenyl, /'so-propenyl, n- butenyl, sec-butenyl, /'so-butenyl, fert-butenyl, n-pentenyl, isopentenyl, neopentenyl, n- hexenyl, 3-methylhexenyl, 2,2- dimethylpentenyl, 2,3-dimethylpentenyl, n-heptenyl, n- octenyl, n-nonenyl, n-decenyl and the like.
As used herein, the term "alkoxy" refers to alkyl-O-, wherein alkyl is defined herein above. Representative examples of alkoxy include, but are not limited to, methoxy, ethoxy, propoxy, 2-propoxy, butoxy, fert-butoxy, pentyloxy, hexyloxy, cyclopropyloxy-, cyclohexyloxy- and the like. Typically, alkoxy groups have about 1-7, more preferably about 1-4 carbons.
As used herein, the term "cycloalkyl" refers to saturated or unsaturated monocyclic, bicyclic or tricyclic hydrocarbon groups of 3-12 carbon atoms. Unless otherwise provided, cycloalkyl refers to cyclic hydrocarbon groups having between 3 and 9 ring carbon atoms or between 3 and 7 ring carbon atoms. Exemplary monocyclic hydrocarbon groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl and cyclohexenyl and the like. Exemplary bicyclic hydrocarbon groups include bornyl, indyl, hexahydroindyl, tetrahydronaphthyl, decahydronaphthyl, bicyclo[2.1.1]hexyl, bicyclo[2.2.1]heptyl, bicyclo[2.2.1]heptenyl, 6,6- dimethylbicyclo[3.1.1]heptyl, 2,6,6-trimethylbicyclo[3.1.1]heptyl, bicyclo[2.2.2]octyl and the like. Exemplary tricyclic hydrocarbon groups include adamantyl and the like.
As used herein, the term "azacycloalkane" refers to saturated or unsaturated monocyclic, bicyclic or tricyclic hydrocarbon groups of 3-12 carbon atoms as defined for "cycloalkyl", wherein one carbon atom is replaced by a nitrogen atom. Unless otherwise provided, azacycloalkyi refers to cyclic aza-hydrocarbon groups having between 2 and 9 ring carbon atoms and one nitrogen atom or between 2 and 7 ring carbon atoms and one nitrogen atom. Exemplary monocyclic aza-hydrocarbon groups include, but are not limited to, aziridinyl, azetidinly, pyrollidinyl, piperidinyl, azepanyl, dihydroazepinyl and the like.
As used herein, the term "halogen" or "halo" refers to fluoro, chloro, bromo, and iodo.
As used herein the term "heterocyclic", "heterocyclyl" or "heterocyclo" may refer to a saturated or unsaturated non-aromatic ring or ring system, e.g., which is a 4-, 5-, 6-, or 7-membered monocyclic, 7-, 8-, 9-, 10-, 1 1-, or 12-membered bicyclic or 10-, 1 1-, 12-, 13-, 14- or 15-membered tricyclic ring system and contains at least one heteroatom selected from O, S and N, where the N and S can also optionally be oxidized to various oxidation states. The heterocyclic group can be attached at a heteroatom or a carbon atom. The heterocyclyl may include fused or bridged rings as well as spirocyclic rings. Examples of heterocycles include azetidine, tetrahydrofuran (THF), dihydrofuran, 1 , 4- dioxane, morpholine, 1 ,4-dithiane, piperazine, piperidine, 1 ,3-dioxolane, imidazolidine, imidazoline, pyrroline, pyrrolidine, tetrahydropyran, dihydropyran, oxathiolane, dithiolane, 1 ,3-dioxane, 1 ,3-dithiane, oxathiane, thiomorpholine, and the like.
As used herein, the term "aryloxy" refers to both an -O-aryl and an -O-heteroaryl group, wherein aryl and heteroaryl are defined herein.
As used herein, the term "heteroaryl" refers to a 5-14 membered monocyclic- or bicyclic- or tricyclic-aromatic ring system, having 1 to 8 heteroatoms selected from N, O or S. Typically, the heteroaryl is a 5-10 membered ring system (e.g., 5-7 membered monocycle or an 8-10 memberred bicycle) or a 5-7 membered ring system. Typical heteroaryl groups include 2- or 3-thienyl, 2- or 3-furyl, 2- or 3-pyrrolyl, 2-, 4-, or 5- imidazolyl, 3-, 4-, or 5- pyrazolyl, 2-, 4-, or 5-thiazolyl, 3-, 4-, or 5-isothiazolyl, 2-, 4-, or 5- oxazolyl, 3-, 4-, or 5-isoxazolyl, 3- or 5-1 ,2,4-triazolyl, 4- or 5-1 ,2, 3-triazolyl, tetrazolyl, 2- , 3-, or 4-pyridyl, 3- or 4-pyridazinyl, 3-, 4-, or 5-pyrazinyl, 2-pyrazinyl, and 2-, 4-, or 5- pyrimidinyl.
The term "heteroaryl" also refers to a group in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where the radical or point of attachment is on the heteroaromatic ring. Nonlimiting examples include 1-, 2-, 3-, 5-, 6-, 7-, or 8- indolizinyl, 1-, 3-, 4-, 5-, 6-, or 7-isoindolyl, 2-, 3-, 4-, 5-, 6-, or 7-indolyl, 2-, 3- , 4-, 5-, 6-, or 7-indazolyl, 2-, 4-, 5-, 6-, 7-, or 8- purinyl, 1-, 2-, 3-, 4-, 6-, 7-, 8-, or 9- quinolizinyl, 2-, 3-, 4-, 5-, 6-, 7-, or 8-quinoliyl, 1-, 3-, 4-, 5-, 6-, 7-, or 8-isoquinoliyl, 1-, 4-, 5-, 6-, 7-, or 8-phthalazinyl, 2-, 3-, 4-, 5-, or 6-naphthyridinyl, 2-, 3- , 5-, 6-, 7-, or 8- quinazolinyl, 3-, 4-, 5-, 6-, 7-, or 8-cinnolinyl, 2-, 4-, 6-, or 7-pteridinyl, 1-, 2-, 3-, 4-, 5-, 6-, 7-, or 8-4aH carbazolyl, 1-, 2-, 3-, 4-, 5-, 6-, 7-, or 8-carbzaolyl, 1-, 3-, 4-, 5-, 6-, 7-, 8-, or 9-carbolinyl, 1-, 2-, 3-, 4-, 6-, 7-, 8-, 9-, or 10-phenanthridinyl, 1- , 2-, 3-, 4-, 5-, 6-, 7-, 8-, or 9-acridinyl, 1-, 2-, 4-, 5-, 6-, 7-, 8-, or 9-perimidinyl, 2-, 3-, 4-, 5-, 6-, 8-, 9-, or 10- phenathrolinyl, 1-, 2- , 3-, 4-, 6-, 7-, 8-, or 9-phenazinyl, 1-, 2-, 3-, 4-, 6-, 7-, 8-, 9-, or 10- phenothiazinyl, 1-, 2-, 3-, 4-, 6-, 7-, 8-, 9-, or 10-phenoxazinyl, 2-, 3-, 4-, 5-, 6-, or I-, 3-, 4-, 5-, 6-, 7-, 8-, 9-, or 10- benzisoqinolinyl, 2-, 3-, 4-, or thieno[2,3-b]furanyl, 2-, 3-, 5-, 6- , 7-, 8-, 9-, 10 -, or 1 1-7H-pyrazino[2,3-c]carbazolyl,2-, 3-, 5-, 6-, or 7-2H- furo[3,2-b]- pyranyl, 2-, 3-, 4-, 5-, 7-, or 8-5H-pyrido[2,3-d]-o-oxazinyl, 1-, 3-, or 5-1 H-pyrazolo[4,3-d]- oxazolyl, 2-, 4-, or 54H-imidazo[4,5-d] thiazolyl, 3-, 5-, or 8-pyrazino[2,3-d]pyridazinyl, 2-,
3- , 5-, or 6- imidazo[2,1-b] thiazolyl, 1-, 3-, 6-, 7-, 8-, or 9-furo[3,4-c]cinnolinyl, 1-, 2-, 3-,
4- , 5-, 6-, 8-, 9-, 10, or 1 1-4H-pyrido[2,3-c]carbazolyl, 2-, 3-, 6-, or 7-imidazo[1 ,2- b][1 ,2,4]triazinyl, 7-benzo[b]thienyl, 2-, 4-, 5- , 6-, or 7-benzoxazolyl, 2-, 4-, 5-, 6-, or 7- benzimidazolyl, 2-, 4-, 4-, 5-, 6-, or 7-benzothiazolyl, 1-, 2-, 4-, 5-, 6-, 7-, 8-, or 9- benzoxapinyl, 2-, 4-, 5-, 6-, 7-, or 8-benzoxazinyl, 1-, 2-, 3-, 5-, 6-, 7-, 8-, 9-, 10-, or 1 1- 1 H-pyrrolo[1 ,2-b][2]benzazapinyl. Typical fused heteroary groups include, but are not limited to 2-, 3-, 4-, 5-, 6-, 7-, or 8-quinolinyl, 1-, 3-, 4-, 5-, 6-, 7-, or 8-isoquinolinyl, 2-, 3-, 4-, 5-, 6-, or 7-indolyl, 2-, 3-, 4-, 5-, 6-, or 7-benzo[b]thienyl, 2-, 4-, 5- , 6-, or 7- benzoxazolyl, 2-, 4-, 5-, 6-, or 7-benzimidazolyl, and 2-, 4-, 5-, 6-, or 7-benzothiazolyl.
As used herein, the terms "salt" or "salts" refers to an acid addition or base addition salt of a compound of the invention. "Salts" include in particular "pharmaceutical acceptable salts". The term "pharmaceutically acceptable salts" refers to salts that retain the biological effectiveness and properties of the compounds of this invention and, which typically are not biologically or otherwise undesirable. In many cases, the compounds of the present invention are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto.
Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids, e.g., acetate, aspartate, benzoate, besylate, bromide/hydrobromide, bicarbonate/carbonate, bisulfate/sulfate, camphorsulfonate, chloride/hydrochloride, chlortheophyllonate, citrate, ethandisulfonate, fumarate, gluceptate, gluconate, glucuronate, hippurate, hydroiodide/iodide, isethionate, lactate, lactobionate, laurylsulfate, malate, maleate, malonate, mandelate, mesylate, methylsulphate, naphthoate, napsylate, nicotinate, nitrate, octadecanoate, oleate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, polygalacturonate, propionate, stearate, succinate, sulfosalicylate, tartrate, tosylate and trifluoroacetate salts.
Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, toluenesulfonic acid, sulfosalicylic acid, and the like. Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
Inorganic bases from which salts can be derived include, for example, ammonium salts and metals from columns I to XII of the periodic table. In certain embodiments, the salts are derived from sodium, potassium, ammonium, calcium, magnesium, iron, silver, zinc, and copper; particularly suitable salts include ammonium, potassium, sodium, calcium and magnesium salts.
Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like. Certain organic amines include isopropylamine, benzathine, cholinate, diethanolamine, diethylamine, lysine, meglumine, piperazine and tromethamine.
The pharmaceutically acceptable salts of the present invention can be synthesized from a basic or acidic moiety, by conventional chemical methods. Generally, such salts can be prepared by reacting free acid forms of these compounds with a stoichiometric amount of the appropriate base (such as Na, Ca, Mg, or K hydroxide, carbonate, bicarbonate or the like), or by reacting free base forms of these compounds with a stoichiometric amount of the appropriate acid. Such reactions are typically carried out in water or in an organic solvent, or in a mixture of the two. Generally, use of non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile is desirable, where practicable. Lists of additional suitable salts can be found, e.g., in "Remington's Pharmaceutical Sciences", 20th ed., Mack Publishing Company, Easton, Pa., (1985); and in "Handbook of Pharmaceutical Salts: Properties, Selection, and Use" by Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002).
Any formula given herein is also intended to represent unlabeled forms as well as isotopically labeled forms of the compounds. Isotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, and chlorine, such as 2H, 3H, 11C, 13C, 14C, 15N, 18F 31P, 32P, 35S, 36CI, 125l respectively. The invention includes various isotopically labeled compounds as defined herein, for example those into which radioactive isotopes, such as 3H and 14C, or those into which non-radioactive isotopes, such as 2H and 13C are present. Such isotopically labelled compounds are useful in metabolic studies (with 14C), reaction kinetic studies (with, for example 2H or 3H), detection or imaging techniques, such as positron emission tomography (PET) or single- photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients. In particular, an 18F or labeled compound may be particularly desirable for PET or SPECT studies. Isotopically-labeled compounds of formula (I) can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically-labeled reagents in place of the non-labeled reagent previously employed.
Further, substitution with heavier isotopes, particularly deuterium (i.e., 2H or D) may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements or an improvement in therapeutic index. It is understood that deuterium in this context is regarded as a substituent of a compound of the formula (I). The concentration of such a heavier isotope, specifically deuterium, may be defined by the isotopic enrichment factor. The term "isotopic enrichment factor" as used herein means the ratio between the isotopic abundance and the natural abundance of a specified isotope. If a substituent in a compound of this invention is denoted deuterium, such compound has an isotopic enrichment factor for each designated deuterium atom of at least 3500 (52.5% deuterium incorporation at each designated deuterium atom), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation).
Pharmaceutically acceptable solvates in accordance with the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D20, de- acetone, d6-DMSO.
Compounds of the invention, i.e. compounds of formula (I) that contain groups capable of acting as donors and/or acceptors for hydrogen bonds may be capable of forming co- crystals with suitable co-crystal formers. These co-crystals may be prepared from compounds of formula (I) by known co-crystal forming procedures. Such procedures include grinding, heating, co-subliming, co-melting, or contacting in solution compounds of formula (I) with the co-crystal former under crystallization conditions and isolating co- crystals thereby formed. Suitable co-crystal formers include those described in WO 2004/078163. Hence the invention further provides co-crystals comprising a compound of formula (I).
As used herein, the term "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, surfactants, antioxidants, preservatives (e.g., antibacterial agents, antifungal agents), isotonic agents, absorption delaying agents, salts, preservatives, drug stabilizers, binders, excipients, disintegration agents, lubricants, sweetening agents, flavoring agents, dyes, and the like and combinations thereof, as would be known to those skilled in the art (see, for example, Remington's Pharmaceutical Sciences, 18th Ed. Mack Printing Company, 1990, pp. 1289- 1329). Except insofar as any conventional carrier is incompatible with the active ingredient, its use in the therapeutic or pharmaceutical compositions is contemplated. The term "a therapeutically effective amount" of a compound of the present invention refers to an amount of the compound of the present invention that will elicit the biological or medical response of a subject, for example, reduction or inhibition of an enzyme or a protein activity, or ameliorate symptoms, alleviate conditions, slow or delay disease progression, or prevent a disease, etc. In one non-limiting embodiment, the term "a therapeutically effective amount" refers to the amount of the compound of the present invention that, when administered to a subject, is effective to (1 ) at least partially alleviating, inhibiting, preventing and/or ameliorating a condition, or a disorder or a disease (i) mediated by Btk, or (ii) associated with Btk activity, or (iii) characterized by activity (normal or abnormal) of Btk; or (2) reducing or inhibiting the activity of Btk; or (3) reducing or inhibiting the expression of Btk. In another non-limiting embodiment, the term "a therapeutically effective amount" refers to the amount of the compound of the present invention that, when administered to a cell, or a tissue, or a non-cellular biological material, or a medium, is effective to at least partially reducing or inhibiting the activity of Btk; or reducing or inhibiting the expression of Btk partially or completely.
As used herein, the term "subject" refers to an animal. Typically the animal is a mammal. A subject also refers to for example, primates (e.g., humans, male or female), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, birds and the like. In certain embodiments, the subject is a primate. In yet other embodiments, the subject is a human.
As used herein, the term "inhibit", "inhibition" or "inhibiting" refers to the reduction or suppression of a given condition, symptom, or disorder, or disease, or a significant decrease in the baseline activity of a biological activity or process.
As used herein, the term "treat", "treating" or "treatment" of any disease or disorder refers in one embodiment, to ameliorating the disease or disorder (i.e., slowing or arresting or reducing the development of the disease or at least one of the clinical symptoms thereof). In another embodiment "treat", "treating" or "treatment" refers to alleviating or ameliorating at least one physical parameter including those which may not be discernible by the patient. In yet another embodiment, "treat", "treating" or
"treatment" refers to modulating the disease or disorder, either physically, (e.g., stabilization of a discernible symptom), physiologically, (e.g., stabilization of a physical parameter), or both. In yet another embodiment, "treat", "treating" or "treatment" refers to preventing or delaying the onset or development or progression of the disease or disorder.
As used herein, a subject is "in need of" a treatment if such subject would benefit biologically, medically or in quality of life from such treatment.
As used herein, the term "a," "an," "the" and similar terms used in the context of the present invention (especially in the context of the claims) are to be construed to cover both the singular and plural unless otherwise indicated herein or clearly contradicted by the context.
All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context. The use of any and all examples, or exemplary language (e.g. "such as") provided herein is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention otherwise claimed.
Any asymmetric atom (e.g. , carbon or the like) of the compound(s) of the present invention can be present in racemic or enantiomerically enriched, for example the (R)-, (S)- or (R,S)- configuration. In certain embodiments, each asymmetric atom has at least 50 % enantiomeric excess, at least 60 % enantiomeric excess, at least 70 %
enantiomeric excess, at least 80 % enantiomeric excess, at least 90 % enantiomeric excess, at least 95 % enantiomeric excess, or at least 99 % enantiomeric excess in the (R)- or (S)- configuration. Substituents at atoms with unsaturated double bonds may, if possible, be present in c/'s- (Z)- or trans- (E)- form.
Accordingly, as used herein a compound of the present invention can be in the form of one of the possible isomers, rotamers, atropisomers, tautomers or mixtures thereof, for example, as substantially pure geometric (c/'s or trans) isomers, diastereomers, optical isomers (antipodes), racemates or mixtures thereof.
Any resulting mixtures of isomers can be separated on the basis of the physicochemical differences of the constituents, into the pure or substantially pure geometric or optical isomers, diastereomers, racemates, for example, by chromatography and/or fractional crystallization. Any resulting racemates of final products or intermediates can be resolved into the optical antipodes by known methods, e.g., by separation of the diastereomeric salts thereof, obtained with an optically active acid or base, and liberating the optically active acidic or basic compound. In particular, a basic moiety may thus be employed to resolve the compounds of the present invention into their optical antipodes, e.g., by fractional crystallization of a salt formed with an optically active acid, e.g., tartaric acid, dibenzoyl tartaric acid, diacetyl tartaric acid, di-0,0'-p-toluoyl tartaric acid, mandelic acid, malic acid or camphor-10-sulfonic acid. Racemic products can also be resolved by chiral chromatography, e.g., high pressure liquid chromatography (HPLC) using a chiral adsorbent.
Furthermore, the compounds of the present invention, including their salts, can also be obtained in the form of their hydrates, or include other solvents used for their crystallization. The compounds of the present invention may inherently or by design form solvates with pharmaceutically acceptable solvents (including water); therefore, it is intended that the invention embrace both solvated and unsolvated forms. The term "solvate" refers to a molecular complex of a compound of the present invention
(including pharmaceutically acceptable salts thereof) with one or more solvent molecules. Such solvent molecules are those commonly used in the pharmaceutical art, which are known to be innocuous to the recipient, e.g., water, ethanol, and the like. The term "hydrate" refers to the complex where the solvent molecule is water.
The compounds of the present invention, including salts, hydrates and solvates thereof, may inherently or by design form polymorphs.
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of the present invention and a pharmaceutically acceptable carrier. The pharmaceutical composition can be formulated for particular routes of administration such as oral administration, parenteral administration, and rectal administration, etc. In addition, the pharmaceutical compositions of the present invention can be made up in a solid form (including without limitation capsules, tablets, pills, granules, powders or suppositories), or in a liquid form (including without limitation solutions, suspensions or emulsions). The pharmaceutical compositions can be subjected to conventional pharmaceutical operations such as sterilization and/or can contain conventional inert diluents, lubricating agents, or buffering agents, as well as adjuvants, such as preservatives, stabilizers, wetting agents, emulsifiers and buffers, etc.
Typically, the pharmaceutical compositions are tablets or gelatin capsules comprising the active ingredient together with a) diluents, e.g., lactose, dextrose, sucrose, mannitol, sorbitol, cellulose and/or glycine; b) lubricants, e.g., silica, talcum, stearic acid, its magnesium or calcium salt and/or polyethyleneglycol; for tablets also c) binders, e.g., magnesium aluminum silicate, starch paste, gelatin, tragacanth, methylcellulose, sodium carboxymethylcellulose and/or polyvinylpyrrolidone; if desired d) disintegrants, e.g., starches, agar, alginic acid or its sodium salt, or effervescent mixtures; and/or e) absorbents, colorants, flavors and sweeteners.
Tablets may be either film coated or enteric coated according to methods known in the art.
Suitable compositions for oral administration include an effective amount of a compound of the invention in the form of tablets, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsion, hard or soft capsules, or syrups or elixirs. Compositions intended for oral use are prepared according to any method known in the art for the manufacture of pharmaceutical compositions and such compositions can contain one or more agents selected from the group consisting of sweetening agents, flavoring agents, coloring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations. Tablets may contain the active ingredient in admixture with nontoxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients are, for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, corn starch, or alginic acid; binding agents, for example, starch, gelatin or acacia; and lubricating agents, for example magnesium stearate, stearic acid or talc. The tablets are uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate can be employed. Formulations for oral use can be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil.
Certain injectable compositions are aqueous isotonic solutions or suspensions, and suppositories are advantageously prepared from fatty emulsions or suspensions. Said compositions may be sterilized and/or contain adjuvants, such as preserving, stabilizing, wetting or emulsifying agents, solution promoters, salts for regulating the osmotic pressure and/or buffers. In addition, they may also contain other therapeutically valuable substances. Said compositions are prepared according to conventional mixing, granulating or coating methods, respectively, and contain about 0.1-75%, or contain about 1-50%, of the active ingredient.
Suitable compositions for transdermal application include an effective amount of a compound of the invention with a suitable carrier. Carriers suitable for transdermal delivery include absorbable pharmacologically acceptable solvents to assist passage through the skin of the host. For example, transdermal devices are in the form of a bandage comprising a backing member, a reservoir containing the compound optionally with carriers, optionally a rate controlling barrier to deliver the compound of the skin of the host at a controlled and predetermined rate over a prolonged period of time, and means to secure the device to the skin.
Suitable compositions for topical application, e.g., to the skin and eyes, include aqueous solutions, suspensions, ointments, creams, gels or sprayable formulations, e.g., for delivery by aerosol or the like. Such topical delivery systems will in particular be appropriate for dermal application, e.g., for the treatment of skin cancer, e.g., for prophylactic use in sun creams, lotions, sprays and the like. They are thus particularly suited for use in topical, including cosmetic, formulations well-known in the art. Such may contain solubilizers, stabilizers, tonicity enhancing agents, buffers and
preservatives. As used herein a topical application may also pertain to an inhalation or to an intranasal application. They may be conveniently delivered in the form of a dry powder (either alone, as a mixture, for example a dry blend with lactose, or a mixed component particle, for example with phospholipids) from a dry powder inhaler or an aerosol spray presentation from a pressurised container, pump, spray, atomizer or nebuliser, with or without the use of a suitable propellant.
The present invention further provides anhydrous pharmaceutical compositions and dosage forms comprising the compounds of the present invention as active ingredients, since water may facilitate the degradation of certain compounds.
Anhydrous pharmaceutical compositions and dosage forms of the invention can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions. An anhydrous pharmaceutical composition may be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions are packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e. g., vials), blister packs, and strip packs.
The invention further provides pharmaceutical compositions and dosage forms that comprise one or more agents that reduce the rate by which the compound of the present invention as an active ingredient will decompose. Such agents, which are referred to herein as "stabilizers," include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers, etc.
The compounds of formula I in free form or in salt form, exhibit valuable pharmacological properties, e.g. Btk modulating properties, e.g. as indicated by in vitro and in vivo tests as provided in the next sections and are therefore indicated for therapy.
Compounds of the invention may be useful in the treatment of an indication selected from: Autoimmune disorders, inflammatory diseases, allergic diseases, airway diseases, such as asthma and chronic obstructive pulmonary disease (COPD), transplant rejection; diseases in which antibody production, antigen presentation, cytokine production or lymphoid organogenesis are abnormal or are undesirable; including rheumatoid arthritis, systemic onset juvenile idiopathic arthritis (SOJIA), gout, pemphigus vulgaris, idiopathic thrombocytopenic purpura, systemic lupus
erythematosus, multiple sclerosis, myasthenia gravis, Sjogren's syndrome, autoimmune hemolytic anemia, anti-neutrophil cytoplasmic antibodies (ANCA)-associated
vasculitides, cryoglobulinemia, thrombotic thrombocytopenic purpura, chronic autoimmune urticaria, allergy (atopic dermatitis, contact dermatitis, allergic rhinitis), atherosclerosis, type 1 diabetes, type 2 diabetes, inflammatory bowel disease, ulcerative colitis, morbus Crohn, pancreatitis, glomerolunephritis, Goodpasture's syndrome, Hashimoto's thyroiditis, Grave's disease, antibody-mediated transplant rejection (AMR), graft versus host disease, B cell-mediated hyperacute, acute and chronic transplant rejection; thromboembolic disorders, myocardial infarct, angina pectoris, stroke, ischemic disorders, pulmonary embolism; cancers of haematopoietic origin including but not limited to multiple myeloma; leukemia; acute myelogenous leukemia; chronic myelogenous leukemia; lymphocytic leukemia; myeloid leukemia; non-Hodgkin lymphoma; lymphomas; polycythemia vera; essential thrombocythemia; myelofibrosis with myeloid metaplasia; and Waldenstroem disease.
Thus, as a further embodiment, the present invention provides the use of a compound of formula (I) or a salt thereof in therapy. In a further embodiment, the therapy is selected from a disease which may be treated by inhibition of Btk. In another embodiment, the disease is selected from the afore-mentioned list, suitably rheumatoid arthritis, systemic onset juvenile idiopathic arthritis (SOJIA), pemphigus vulgaris, idiopathic
thrombocytopenic purpura, systemic lupus erythematosus, multiple sclerosis, myasthenia gravis, Sjogren's syndrome, autoimmune hemolytic anemia, anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitides, cryoglobulinemia, thrombotic thrombocytopenic purpura, chronic autoimmune urticaria, atopic dermatitis, allergic rhinitis, type 1 diabetes, type 2 diabetes, inflammatory bowel disease, morbus Crohn, Goodpasture's syndrome, Grave's disease, antibody-mediated transplant rejection (AMR), B cell-mediated hyperacute, acute and chronic transplant rejection; multiple myeloma; acute myelogenous leukemia; chronic myelogenous leukemia; lymphocytic leukemia; myeloid leukemia; non-Hodgkin lymphoma; myelofibrosis with myeloid metaplasia; and Waldenstroem disease, more suitably rheumatoid arthritis, systemic onset juvenile idiopathic arthritis (SOJIA), systemic lupus erythematosus, multiple sclerosis, Sjogren's syndrome, chronic autoimmune urticaria, atopic dermatitis, allergic rhinitis, type 1 diabetes, type 2 diabetes, inflammatory bowel disease, multiple myeloma; non-Hodgkin lymphoma.
In another embodiment, the invention provides a method of treating a disease which is treated by inhibition of Btk kinase comprising administration of a therapeutically acceptable amount of a compound of formula (I) or a salt thereof. In a further embodiment, the disease is selected from the afore-mentioned list, suitably rheumatoid arthritis, systemic onset juvenile idiopathic arthritis (SOJIA), pemphigus vulgaris, idiopathic thrombocytopenic purpura, systemic lupus erythematosus, multiple sclerosis, myasthenia gravis, Sjogren's syndrome, autoimmune hemolytic anemia, anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitides, cryoglobulinemia, thrombotic thrombocytopenic purpura, chronic autoimmune urticaria, atopic dermatitis, allergic rhinitis, type 1 diabetes, type 2 diabetes, inflammatory bowel disease, morbus Crohn, Goodpasture's syndrome, Grave's disease, antibody-mediated transplant rejection (AMR), B cell-mediated hyperacute, acute and chronic transplant rejection; multiple myeloma; acute myelogenous leukemia; chronic myelogenous leukemia; lymphocytic leukemia; myeloid leukemia; non-Hodgkin lymphoma; myelofibrosis with myeloid metaplasia; and Waldenstroem disease, more suitably rheumatoid arthritis, systemic onset juvenile idiopathic arthritis (SOJIA), systemic lupus erythematosus, multiple sclerosis, Sjogren's syndrome, chronic autoimmune urticaria, atopic dermatitis, allergic rhinitis, type 1 diabetes, type 2 diabetes, inflammatory bowel disease, multiple myeloma; non-Hodgkin lymphoma.
Methods of synthesizing Pyrrolo-pyrimidines
Agents of the invention, i.e. compounds in accordance to the definition of formula (I), may be prepared by a reaction sequence (shown below) involving Suzuki coupling of a protected boronic esters II with the corresponding aryl halides Γ, conveniently furnishing intermediate III. Deprotection of III, e.g. with diluted hydrochloric acid in methanol or the like and acylation, e.g. with an appropriate acetylating agent, e.g. unsubstituted or substituted acetanhydride in the absence or presence of a solvent, of IV is followed by an additional Suzuki coupling with boronic ester VI and acylation of VII (optionally followed by a deprotection step), as shown in Reaction Scheme 1 below, wherein X denotes N, and the group PG refers to a protecting group such as for example, tert- butyloxycarbonyl, that may be easily removed e.g. by diluted hydrochloric acid in methanol or the like.
Reaction Scheme 1 :
Figure imgf000027_0001
Alternatively, the compounds of formula (I) may also be prepared by a reaction sequence involving Suzuki coupling of boronic esters IX with the corresponding aryl halides Γ , followed by an additional Suzuki coupling of V with boronic esters VI and an acylation of VII (optionally followed by a deprotection step), as shown in Reaction Scheme 2 below:
Reaction Scheme 2:
Figure imgf000028_0001
Alternatively, the compounds of formula (I) may also be prepared by a reaction sequence involving Suzuki coupling of boronic esters VI with the corresponding aryl halides III, acylation of intermediate X, followed by deprotection of XI and acylation of XII (optionally followed by a deprotection step), as shown in Reaction Scheme 3 below, wherein X denotes N, and the group PG refers to a protecting group that may be easily removed, such as for example, tert-butyloxycarbonyl.
Reaction Scheme 3:
Figure imgf000029_0001
Figure imgf000029_0002
Figure imgf000029_0003
Alternatively, the compounds of formula (I) may also be prepared by a reaction sequence involving Suzuki coupling of boronic esters XIII with the corresponding aryl halides III, followed by deprotection of XI and acylation of XII (optionally followed by a deprotection step), as shown in Reaction Scheme 4 below, wherein X denotes N, and the group PG refers to a protecting group that may be easily removed, such as for example, tert-butyloxycarbonyl. Reaction Scheme 4:
Figure imgf000030_0001
Alternatively, the compounds of formula (I) may also be prepared by a Suzuki coupling of boronic esters XIV with the corresponding aryl halides III (optionally followed by a deprotection step), as shown in Reaction Scheme 5 below:
Figure imgf000030_0002
Alternatively, the compounds of formula (I) may also be prepared by urea formation reaction of anilines VII (optionally followed by a deprotection step), as shown in Reaction Scheme 6 below:
Reaction Scheme 6:
Figure imgf000031_0001
Alternatively, the compounds of formula (I) may be prepared by a reaction sequence involving Suzuki coupling of boronic esters XIII with the corresponding aryl halides XVI. Protection of XVII is followed by halogenation XVII, deprotection of XIX, and Suzuki coupling of halide XX with boronic ester IX (optionally followed by a deprotection step), as shown in Reaction Scheme 7 below, wherein the group PG refers to a protecting group that may be easily removed, such as for example, benzenesulfonyl.
Reaction Scheme 7:
Figure imgf000032_0001
Synthesis of the compound(s) of the invention:
EXPERIMENTAL SECTION Abbreviations:
AcOH Acetic acid
BOC tert-Butyloxycarbonyl
COMU: (1-Cyano-2-ethoxy-2-oxoethylidenaminooxy)dimethylamino-morpholino- carbenium hexafluorophosphate
Cs2C03 Cesium carbonate
DCM: Dichloromethane
DIPEA: Ethyl-diisopropyl-amine, Hunig's base, DIEA
DMA: N,N-Dimethylacetamide
DMAP: Dimethyl-pyridin-4-yl-amine
DMF: Ν,Ν-Dimethyl formamide
DMSO: Dimethylsulfoxide
EtOAc: Acetic acid ethyl ester
EtOH: Ethanol
HATU : 0-(7-Azabenzotriazol- 1 -yl)-N , N , N ' , N '- tetramethyluroniumhexafluorophosphat
hrs: Hours
LDA: Lithium diisopropyamide
MeCN: Acetonitrile
MeOH: Methanol
NaBH4: Sodium borohydride
NaH sodium hydride
Na2S04 Sodium sulfate
NH4OH: Ammonia hydrogen solution 25 %
Pd/C: Palladium on carbon
Pd2(dba)3: Tn's(dibenzylideneacetone)dipalladium(0)
TEA: Triethylamine
TFA: Trifluoro-acetic acid THF: Tetrahydrofuran
rt: Retention time
r.t. Room temperature
Xantphos: 4,5-Bis-diphenylphosphanyl-9,9-dimethyl-9H-xanthene
1H-NMR spectra were recorded on a Bruker 600 MHz, a Bruker 500 MHz, or a Bruker 400 MHz NMR spectrometer. Significant peaks are tabulated in the order: multiplicity (s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; br, broad) and number of protons. Electron Spray Ionization (ESI) mass spectra were recorded on an Agilent 1 100 Series mass spectrometer. Mass spectrometry results are reported as the ratio of mass over charge.
Detailed analytical HPLC chromatography methods referred to in the preparations and examples below are outlined as follows:
Preparative LC/MS Method 1 :
Preparative Waters chromatography instrument equipped with a mircomass ZQ MS detector and Waters X bridge C18-ODB (5 μηη) 30x150mm column. Peak detection is reported at 210 nm wavelength.
Solvent A: Water containing 1 mM ammonium hydrogen carbonate
Solvent B: Acetonitrile containing 0.04% formic acid.
Flow rate at 50 ml/minute
Gradient: Time [minutes] Solvent A [%] Solvent B [%]
0 85 15
1 85 15
10 10 80
1 1 0 100
13.5 0 100
LC/MS Method 1 :
Waters Acquity UPLC instrument equipped with diode array detector, Waters SQD Single Stage Quadrupole mass spectrometer and Waters Acquity HSS T3 (1.8μηη) 2.1x50mm column. Peak detection is reported full scan 210 - 315 nm wavelength. Column temperature 50 C.
Solvent A: Water containing 0.05% ammonium acetate and 0.05% formic acid. Solvent B: Acetonitrile containing 0.04% formic acid.
Flow rate at 1.2 ml/minute
Gradient: Time [minutes] Solvent A [%] Solvent B [%]
0 98 2
1.40 2 98
2.15 2 98
2.19 98 2
2.20 98 2
Mass range: ESI +/-: 120 - 1200 m/z
All reagents, starting materials and intermediates utilized in these examples are available from commercial sources or are readily prepared by methods known to those skilled in the art.
Example 1
4-(4-(5-Fluoro-3-r4-(1-fluoro-cvclopropyl)-benzoylaminol-2-methyl-phenyl)-7H- Pyrrolor2 -dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000036_0001
(1 ) 4-(4-Chloro-7H-pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid tert-butyl ester, Intermediate 1
Figure imgf000036_0002
To a mixture of 4-chloro-6-iodo-7H-pyrrolo[2,3-d]pyrimidine (2.6 g, 9.30 mmol) and dichlorobis(triphenylphosphine)palladium (II) (0.52 g, 0.74 mmol) in 1-propanol (120 ml) and aqueous sodium carbonate solution (2M, 10.23 ml, 20.46 mmol), 4-(4, 4,5,5- tetramethyl-[1 ,2,3]dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester (3.02 g, 9.77 mmol) was added. The mixture was heated to 100°C for 18 hours. After cooling the brownish mixture was diluted with 200 ml water and extracted with DCM. The organic layer was washed with brine (2x) and dried over sodium sulfate, than filtered and evaporated. The residue was purified by flash chromatography on silica (cyclohexane/EtOAc 1 :1 ) to afford the compound Intermediate 1 as a beige solid.
MS (ESI): 335 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.64 (br s, 1 H), 8.56 (s, 1 H), 6.59 (s, 2H), 4.08 (br s, 2H), 3.57 (m, 2H), 2.55 (m, 2H), 1.45 (s, 9H). (2) 4-Chloro-6-(1.2.3.6-tetrahvdro-pyridin-4-vn-7H-pyrrolor2.3-dlpyrimidine.
Intermediate 2
Figure imgf000037_0001
To a solution of compound Intermediate 1 (2.60 g, 7.77 mmol) in 10 ml DCM, 30 ml TFA/water 95:5 were added. The resulting mixture was stirred at room temperature for 2 hours. The mixture was diluted with water and basified by addition of 2N sodium hydroxide and extracted with DCM. The organic layer was washed with brine (2x), dried over sodium sulfate and evaporated. The residue was purified by flash chromatography on silica (cyclohexane/EtOAc 1 :1 to EtOAc) to afford the compound Intermediate 2 as a beige solid.
MS (ESI): 235 [M+H]+, 1H-NMR (DMSO-d6): δ (ppm) 9.70 (br s, 2H), 8.56 (s, 1 H), 6.58 (s, 1 H), 6.55 (br s, 1 H), 3.80 (m, 2H), 3.30 (m, 2H), 2.80 (m, 2H).
(3) 4-(4-Chloro-6-(1.2.3.6-tetrahvdro-pyridin-4-vn-7H-pyrrolor2.3-dlpyrimidine.
Intermediate 3
Figure imgf000037_0002
To a suspension of the amine Intermediate 2 (0.75 g, 3.20 mmol) in DCM (30 ml) and DIPEA (1.09 ml, 6.39 mmol), dimethylcarbamoylchloride (0.52 g, 4.79 mmol) was added. The mixture was stirred at room temperature for 20 hours. The solvents were evaporated under reduced pressure. The residue was purified by flash chromatography on silica (EtOAc to EtOAc/methanol/ammonia 95:5:0.5) to afford the compound Intermediate 3 as a beige solid.
MS (ESI): 306 [M+H]+, 1H-NMR (DMSO-d6): δ (ppm) 12.65 (s, 1 H), 8.55 (s, 1 H), 6.60 (s, 1 H), 6.57 (s, 1 H), 3.90 (m, 2H), 3.36 (m, 2H), 2.77 (s, 6H), 2.57 (m, 2H). (4) 2-(Fluoro-2-methyl-3-nitro-phenyl)-4,4,5,5-tetramethyl-ri ,3,21dioxaborolone, Intermediate 4
Figure imgf000038_0001
To a mixture of 1-Bromo-5-fluoro-2-methyl-3-nitro-benzene (5.0 g, 21.37 mmol) and bis(diphenylphosphino)ferrocenedichloropalladium (II) (0.78 g, 1.06 mmol) in 200 ml dioxane, bis-(pinacolato)-diboron (8.14 g, 32.0 mmol) and potassium acetate (7.34 g, 74.8 mmol) were added. The mixture was heated to 100°C for 6 hours. After cooling the brownish mixture was diluted with 200 ml water and extracted with EtOAc. The organic layer was washed with sodium hydrogen carbonate (1x) and brine (2x) and dried over sodium sulfate, then filtered and evaporated. The residue was purified by flash chromatography on silica (cyclohexane/EtOAc 9: 1 ) to afford the compound Intermediate 4 as a yellow oil.
MS (ESI): 281 [M]+ , 1H-NMR (DMSO-d6): δ (ppm) 7.79 (d, 1 H), 7.55 (d, 1 H), 2.48 (s, 3H), 1.31 (s, 12H).
(5) 5-Fluoro-2-methyl-3-(4,4,5,5-tetramethyl-ri ,3,2ldioxaborolan-2-yl)-phenylamine, Intermediate 5
Figure imgf000038_0002
The nitro compound Intermediate 4 (12.4 g, 44.1 mmol) was dissolved in 300 ml EtOAc and Pd/C 10% (Pd) (4.0 g) was added. The mixture was hydrogenated at room temperature and normal pressure for 18 hours. The mixture was filtered over Kieselgur (Supelco) and evaporated. The residue was purified by flash chromatography on silica (EtOAc) to afford the compound Intermediate 5 as a beige solid. MS (ESI): 252 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 6.52 (m, 2H), 5.11 (br s, 2NH), 2.19 (s, 3H), 1.29 (s, 12H).
(6) 4-r4-(3-Amino-5-fluoro-2-methyl-p^
dihvdro-2H-pyridine-1-carboxylic acid dimethylamide, Intermediate 6
Figure imgf000039_0001
To a mixture of chloro compound Intermediate 3 (3.94 g, 15.70 mmol) and dichlorobis(triphenylphosphine)palladium (II) (0.73 g, 1.04 mmol) in 1-propanol (150 ml) and aqueous sodium carbonate solution (2M, 6.54 ml, 13.08 mmol), boronic ester compound Intermediate 5 (4.0 g, 13.08 mmol) was added. The mixture was heated at 100°C for 16 hours. After cooling the brownish mixture was diluted with 200 ml water and extracted with EtOAc. The organic layer was washed with brine (2x) and dried over sodium sulfate, than filtered and evaporated. The residue was purified by flash chromatography on silica (EtOAc/methanol/ammonia 98:2:0.2 to EtOAc/MeOH/N H40H 9: 1 :0.1 ) to afford the compound Intermediate 6 as a beige solid.
MS (ESI): 395 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 1 1.80 (br s, 1 H), 8.87 (s, 1 H), 6.70 (m, 1 H), 6.55 (m, 1 H), 6.48 (br s, 1 H), 6.38 (br s, 1 H), 4.05 (br s, 2H), 3.90 (br s, 2NH), 3.55 (m, 2H), 2.90 (s, 6H), 2.65 (m, 2H), 2.10 (s, 3H).
(7) 4-(4-(5-Fluoro-3-r4-(1-fluoro-cvclopropyl)-benzoylaminol-2-methyl-phenyl}-7H- Pyrrolor2'3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000039_0002
Anilino compound Intermediate 6 (80 mg, 0.203 mmol), 4-(1-fluoro-cyclopropyl)-benzoic acid (for preparation see WO 07/090752) (40 mg, 0.223 mmol) and COMU (130 mg, 0.304 mmol) were dissolved in DIPEA (0.071 ml, 0.406 mmol) and DMA (1.0 ml). The solution was stirred at room temperature for 18 hrs. The reaction was diluted with 10 ml EtOAc and extracted with water by a Hamilton extractor. The water layer was extracted with EtOAc (5x). The combined organic layers were dried with sodium sulfate and evaporated. The residue was purified by a preparative LC/MS method 1 to afford the compound Example 1 as a beige solid.
MS (ESI): 557 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.43 (br s, 1 H), 10.07 (s, 1 H), 8.83 (s, 1 H), 8.03 (d, 2H), 7.49 (m, 1 H), 7.43 (d, 2H), 7.23 (m, 1 H), 6.59 (br s, 1 H), 6.33 (s, 1 H), 3.90 (m, 2H), 3.33 (m, 2H), 2.77 (s, 6H), 2.54 (m, 2H), 2.12 (s, 3H), 1.55 (m, 2H), 1.26 (m, 2H).
The following Examples 2 - 14 were prepared as outlined in Reaction Scheme 1 and described for Example 1 using the intermediate Intermediate 6 and the appropriate benzoic acid as starting materials.
Example 2
4-(4-f3-r(3,3-Dimethyl-2,3-dihvdro-benzofuran-6-carbonyl)-aminol-5-fluoro-2-methyl- phenyl)-7H-pyrrolor2'3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000040_0001
LC/MS (Method 1 ): rt: 1.01 minutes, MS (ESI): 569 [M+H]+ Example 3
4-(4-{5-Fluoro-2-methyl-3-r(5-methyl-4,5,6,7-tetrahydro
aminol-phenyl)-7H-pyrrolor2 -dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxyN acid dimethylamide
Figure imgf000041_0001
LC/MS (Method 1 ): rt: 1.14 minutes, MS (ESI): 573 [M+H]+ Example 4
4-(4-f5-Fluoro-3-r4-isopropyl-methyl-amino)-benzoylaminol-2-methyl-phenyl)-7H- Pyrrolor2'3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000041_0002
LC/MS (Method 1 ): rt: 1.05 minutes, MS (ESI): 570 [M+H]+ Example 5
3-MethyM H-indole-6-carboxylic acid (3-r6-(1-dimethylcarbamoyl-1 ,2,3,6-tetrahydro- Pyridin-4-yl)-7H-pyrrolor2,3-dlpyrimidin-4-yll-5-fluoro-2-methyl-phenyl)-amide
Figure imgf000042_0001
LC/MS (Method 1 ): rt: 0.95 minutes, MS (ESI): 552 [M+H]+ Example 6
4-(4-f5-Fluoro-3-r4-(2-hvdroxy-1 ,1-dimethyl-ethvn-benzoylaminol-2-methyl-phenyl)-7H- Pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000042_0002
LC/MS (Method 1 ): rt: 0.88 minutes, MS (ESI): 571 [M+H]+
Example 7
4-{4-r5-Fluoro-2-methyl-3-(4-piperidin-1-yl-benzoylamino)-phenyll-7H-pyrrolor2,3- dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000043_0001
LC/MS (Method 1 ): rt: 1.07 minutes, MS (ESI): 582 [M+H]+ Example 8
4-f4-r5-Fluoro-3-(isopropenyl-benzoylamino)-2-methyl-phenyll-7H-pyrrolor2,3- dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000043_0002
LC/MS (Method 1 ): rt: 1.04 minutes, MS (ESI): 539 [M+H]+
Example 9
4-(4-{5-Fluoro-2-methyl-3-r4-(1-trifluoromethyl-cvclopropyl)-benzoylaminol-phenyl)-7H- Pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000044_0001
LC/MS (Method 1 ): rt: 1.08 minutes, MS (ESI): 607 [M+H]+ Example 10
4-{4-r5-Fluoro-3-(4-isopropoxy-benzoylamino)-2-methyl-phenyll-7H-pyrrolor2,3- dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000044_0002
LC/MS (Method 1 ): rt: 1.04 minutes, MS (ESI): 557 [M+H]+
Example 11
4-(4-{5-Fluoro-3-r4-pentafluorothio-benzoylaminol-2-methyl-phenyl)-7H-pyrrolor2,3- dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000045_0001
LC/MS (Method 1 ): rt: 1.07 minutes, MS (ESI): 625 [M+H]+ Example 12
4-(4-(5-Fluoro-3-r4-(2-methoxy-1 ,1 -dimethyl-ethyl )-benzoylaminol-2-methyl-phenyl)-7H- Pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000045_0002
LC/MS (Method 1 ): rt: 1.04 minutes, MS (ESI): 585 [M+H]+
Example 13
4-(4-{5-Fluoro-3-r4-(1-methoxy-1 -methyl-ethyl )-benzoylaminol-2-methyl-phenyl)-7H- Pyrrolor2,3-dlpyrimidin-6-vn-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000046_0001
LC/MS (Method 1 ): rt: 0.98 minutes, MS (ESI): 571 [M+H]+ Example 14
1-Methyl-1 H-pyrrolor2,3-blpyridine-6-carboxylic acid (3-r6-(1-dimethyl-carbamoyl-1 ,2,3,6- tetrahvdro-pyridin-4-yl)-7H-pyrrolor2,3-dlpyrimidin-4-yll-5-fluoro-2-methyl-phenyl)-amide
Figure imgf000046_0002
LC/MS (Method 1 ): rt: 0.85 minutes, MS (ESI): 553 [M+H]+
Example 15
4-{4-r3-(4-Dimethylamino-benzoylamino)-5-fluoro-2-methyl-phenyll-7H-p
dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000047_0001
To a solution of aniline Intermediate 6 (84 mg, 0.213 mmol) in DCM/pyridine (2:1 , 30 ml) were added DIPEA (0.372 ml, 2.13 mmol), 4-dimethylaminobenzoyl chloride (43 mg, 0.234 mmol), and DMAP (2.6 mg, 0.021 mmol). The resulting mixture was stirred at room temperature for 75 hrs. The solvents were removed in vacuo and the resulting mixture was purified by reversed phase HPLC (MeCN/H20) to afford Example 15.
MS (ESI): 542 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.41 (br s, 1 H), 9.63 (s, 1 H), 8.81 (s, 1 H), 7.88 (d, 2H), 7.48 (m, 1 H), 7.16 (m, 1 H), 6.77 (d, 2H), 6.58 (br s, 1 H), 6.32 (s, 1 H), 3.89 (br s, 2H), 3.32 (m, 2H), 3.00 (s, 6H), 2.76 (s, 6H), 2.54 (m, 2H), 2.10 (s, 3H).
Example 16
4-(4-(3-r2-Fluoro-4-(1 -hydroxy- 1 -methyl-ethyl )-benzoylaminol-2-hvdroxymethyl-phenyl)-
7H-pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000048_0001
(1 ) 4-(4-r3-Amino-2-(tert-butyl-diphenyl-silanyloxymethyl)-phenyll-7H-pyrro dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide, Intermediate 7
Figure imgf000048_0002
Intermediate 7 was prepared analogue to Intermediate 6 by replacing the boronic ester Intermediate 5 with the boronic ester Intermediate 27.
MS (ESI): 631 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.24 (br s, 1 H), 8.52 (s, 1 H), 7.32 (m, 2H), 7.28-7.27 (m, 4H), 716-7.15 (m, 5H), 6.90 (d, 1 H), 6.71 (d, 1 H), 6.53 (m, 1 H), 6.16 (s, 1 H), 5.28 (br s, 2H), 4.82 (s, 2H), 3.88 (m, 6H), 3.31 (m, 2H), 2.75 (s, 6H), 2.44 (m, 2H), 0.81 (s, 9H). i21 2-Fluoro-4-(1-hvdroxy-1-methyl-ethyl)-benzoic acid, Intermediate 8
Figure imgf000048_0003
Intermediate 8 was prepared analogue to Intermediate 35 by replacing Intermediate
34 with 2-fluoro-terephthalic acid 4-methyl ester (J. Med. Chem., 52(19), 5950-5966; 2009).
MS (ESI): 197 [M-H]" , 1H-NMR (DMSO-d6): δ (ppm) 7.80 (t, 1 H), 7.35 (m, 1 H), 7.32 (m, 1 H), 5.27 (br s, 1 H), 1.42 (s, 6H).
(3) 4-(4-f2-(tert-Butyl-diphenyl-silanyloxymethyl)-3-r2-fluoro-4-(1-hydroxy-1 -methyl- ethyl )-benzoylaminol-phenyl)-7H-pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1- carboxylic acid dimethylamide, Intermediate 9
Figure imgf000049_0001
To a mixture of Intermediate 7 (58 mg, 0.091 mmol), Intermediate 8 (27 mg, 0.137 mmol), and DIPEA (0.059 ml, 0.337 mmol) in DMF (10 ml) was added HATU (38 mg, 0.100 mmol). The resulting mixture was stirred at r.t. for 22 hrs. The mixture was diluted with EtOAc and sat. NaHC03 solution. The organic layer was washed with brine and dried over Na2S04. The solvents were removed in vacuo, and the crude product was purified by reversed pahse HPLC (H20/MeCN) to afford Intermediate 9.
(4) 4-(4-f3-r2-Fluoro-4-(1-hvdroxy-1-methyl-ethyl)-benzoylaminol-2-hvdroxymethyl- phenyl)-7H-pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1 -carboxylic acid dimethylamide
Figure imgf000050_0001
Example 16 was prepared analogue to Example 37 step 8 by replacing Intermediate 31 with Intermediate 9.
LC/MS (Method 1 ): rt: 1.94 minutes, MS (ESI): 573 [M+H]+ Example 17
4-(4-r3-(4-Cvclopropyl-benzoylamino)-2-hvdroxymethyl-phenyll-7H-pyrrolor2,3- dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000050_0002
Example 17 was prepared analogue to Example 16 by replacing Intermediate 8 in step 3 with 4-cyclopropylbenzoic acid.
LC/MS (Method 1 ): rt: 2.32 minutes, MS (ESI): 537 [M+H]+ Example 18
4-(4-{5-Fluoro-2-methyl-3-r4-(2,2,2-trifluoro-1-hydroxy-1 -methyl-ethyl Vbenzoylami phenyl)-7H-pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxyli^ acid dimethylamide
Figure imgf000051_0001
Example 18 was prepared analogue to Intermediate 9 by replacing Intermediate 7 with Intermediate 6 and Intermediate 8 with 4-(2,2,2-trifluoro-1-hydroxy-1-methyl-ethyl)- benzoic acid (WO2007/145834).
MS (ESI): 61 1 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.43 (br s, 1 H), 10.10 (s, 1 H), 8.83 (s, 1 H), 8.03 (d, 2H), 7.77 (d, 2H), 7.49 (m, 1 H), 7.22 (m, 1 H), 6.77 (s, 1 H), 6.60 (br s, 1 H), 6.34 (s, 1 H), 3.90 (m, 2H), 3.35 (m, 2H), 2.77 (s, 6H), 2.55 (m, 2H), 2.10 (s, 3H), 1.75 (s, 3H).
Example 19
4-(4-r3-(4-Acetyl-benzoylamino)-5-fluoro-2-methyl-phenyll-7H-pyrrolor2,3-dlpyrimidin-6- yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000051_0002
Example 19 was prepared analogue to Example 18 by replacing 4-(2,2,2-trifluoro-1- hydroxy-1-methyl-ethyl)-benzoic acid with 4-acetyl-benzoic acid.
MS (ESI): 541 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.42 (br s, 1 H), 10.24 (s, 1 H), 8.82 (s, 1 H), 8.1 1 (m, 4H), 7.48 (m, 1 H), 7.24 (m, 1 H), 6.59 (s, 1 H), 6.33 (s, 1 H), 3.89 (m, 2H), 3.34 (m, 2H), 2.76 (s, 6H), 2.65 (s, 3H), 2.53 (m, 2H), 2.13 (s, 3H).
Example 20
4-(4-r3-(4-Cvclopropyl-benzoylamino)-4-fluoro-phen
3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000052_0001
(1 ) 4-r4-(3-Amino-4-fluoro-phenyl)-7H-pyrrolor2,3-dlpyrimidin-6-yll-3,6-dihvdro-2H- pyridine-1-carboxylic acid dimethylamide, Intermediate 10
Figure imgf000052_0002
Intermediate 10 was prepared analogue to Intermediate 6 by replacing Intermediate 5 with 3-amino-4-fluoroboronic acid.
MS (ESI): 381 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.32 (br s, 1 H), 8.74 (s, 1 H), 7.68 (m, 1 H), 7.39 (m, 1 H), 7.13 (m, 1 H), 6.84 (s, 1 H), 6.58 (s, 1 H), 5.36 (br s, 2H), 3.91 (s, 2H), 3.37 (m, 2H), 2.77 (s, 6H), 2.62 (m, 2H). (2) 4-f4-r3-(4-Cvclopropyl-benzoylamino)-4-fluoro-phenyll-7H-pyrrolor2,3- dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000053_0001
A mixture of Intermediate 10 (80mg, 0.210 mmol), 4-cyclopropyl-benzoyl chloride (J. Med. Chem., 52(14), 4329-4337; 2009) (76 mg, 0.421 mmol), and DMAP (2.6 mg, 0.021 mmol) in pyridine (2 ml) was stirred at r.t. overnight. The mixture was evaporated to dryness, then sat. aqueous NaHC03 was added, and the mixture was extracted with DCM. The organic layer was dried, filtered, and evaporated to dryness. The residue was purified by flash chromatography (silica gel, EtOAc/MeOH/NH3 gradient) to obtain Example 20.
MS (ESI): 525 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.44 (br s, 1 H), 10.20 (s, 1 H), 8.80 (s, 1 H), 8.47 (d, 1 H), 8.09 (m, 1 H), 7.92 (d, 2H), 7.49 (t, 1 H), 7.25 (d, 1 H), 6.93 (s, 1 H), 6.60 (s, 1 H), 3.91 (m, 2H), 3.35 (m, 2H), 2.77 (s, 6H), 2.62 (m, 2H), 2.02 (m, 1 H), 1.04 (m, 2H), 0.78 (m, 2H).
Example 21
4-(4-f4-Fluoro-3-r4-(2-hvdroxy-1 , 1-dimethyl-ethyl)-benzoylaminol-phenyl)-7H-pyrrolor2,3- dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000053_0002
Example 21 was prepared analogue to Intermediate 9 by replacing Intermediate 7 with Intermediate 10 and Intermediate 8 with 4-(2-hydroxy-1 ,1-dimethyl-ethyl)-benzoic acid. MS (ESI): 557 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.41 (br s, 1 H), 10.21 (s, 1 H), 8.80 (s, 1 H), 8.50 (m, 1 H), 8.10 (m, 1 H), 7.94 (d, 2H), 7.54 (d, 2H), 7.48 (m, 1 H), 6.93 (s, 1 H), 6.60 (s, 1 H), 4.74 (br s, 1 H), 3.91 (m, 2H), 3.48 (m, 2H), 3.34 (m, 2H), 2.77 (s, 6H), 2.62 (m, 2H). 1.27 (s, 6H).
Example 22
N-(3-r6-(3.6-Dihvdro-2H-pyran-4-vn-7H-pyrrolor2.3-dlpyrimidin-4-yl1-5-fluoro-2-methyl- phenyl)-2-fluoro-4-(1-hydroxy-1 -methyl-ethyl Vbenzamide
Figure imgf000054_0001
(1 ) 4-Chloro-6-(3.6-dihvdro-2H-pyran-4-vn-7H-pyrrolor2.3-dlpyrimidine.
Intermediate 11
Figure imgf000054_0002
Intermediate 11 was prepared analogue to Intermediate 1 by replacing 4-(4, 4,5,5- tetramethyl-[1 ,2,3]dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester with 3,6-dihydro-2H-pyran-4-boronic acid pinacolester.
MS (ESI): 236 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.69 (br s, 1 H), 8.56 (s, 1 H), 6.67 (m, 1 H), 6.58 (s, 1 H), 4.29 (m, 2H), 3.84 (m, 2H), 2.50 (m, 2H). (2) 3-r6-(3.6-Dihvdro-2H-pyran-4-vn-7H-pyrrolor2.3-dlpyrimidin-4-yl1-5-fluoro-2- methyl-phenylamine, Intermediate 12
Figure imgf000055_0001
Intermediate 12 was prepared analogue to Intermediate 6 by replacing Intermediate 3 with Intermediate 11.
MS (ESI): 325 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.34 (br s, 1 H), 8.77 (s, 1 H), 6.62 (m, 1 H), 6.56 (m, 1 H), 6.37 (m, 1 H), 6.25 (s, 1 H), 5.37 (br s, 2H), 4.27 (m, 2H), 3.80 (m, 2H), 2.45 (m, 2H), 1.89 (s, 3H).
(3) N-(3-r6-(3.6-Dihvdro-2H-pyran-4-vn-7H-pyrrolor2.3-dlpyrimidin-4-yl1-5-fluoro-2- methyl-phenyl)-2-fluoro-4-(1-hvdroxy-1-methyl-ethvn-benzamide
Figure imgf000055_0002
Example 22 was prepared analogue to Intermediate 9 by replacing Intermediate 7 with Intermediate 12.
MS (ESI): 505 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.45 (br s, 1 H), 9.95 (s, 1 H), 8.83 (s, 1 H), 7.73 (m, 1 H), 7.63 (m, 1 H), 4.43 (m, 2H), 7.19 (m, 1 H), 6.65 (s, 1 H), 6.34 (s, 1 H), 5.32 (s, 1 H), 4.29 (s, 2H), 3.81 (m, 2H), 2.47 (m, 2H), 2.15 (s, 3H), 1.46 (s, 6H). Example 23
N-(3-r6-(3,6-Dihvdro-2H-pyran-4-yl)-7H-pyrrolor2,3-dlpyrimidin-4-yll-2
phenyl)-2-fluoro-4-(1-hydroxy-1 -methyl-ethyl Vbenzamide
Figure imgf000056_0001
(1 ) 2-(tert-Butyl-diphenyl-silanyloxymethvn-3-r6-(3.6-dihvdro-2H-pyran-4-vn-7H- pyrrolor2,3-dlpyrimidin-4-yll-phenylamine, Intermediate 13
Intermediate 13 was prepared analogue to Intermediate 7 by replacing Intermediate 6 with Intermediate 11.
MS (ESI): 561 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.25 (br s, 1 H), 8.52 (s, 1 H), 7.40- 7.10 (m, 1 1 H), 6.90 (d, 1 H), 6.71 (d, 1 H), 6.59 (s, 1 H), 6.16 (s, 1 H), 5.28 (s, 2H), 4.81 (s, 2H), 4.27 (m, 2H), 3.78 (m, 2H), 2.37 (m, 2H), 0.82 (s, 9H). (2) N-(2- ert-Butyl-diphenyl-silanyloxymethvn-3-r6-(3.6-dihvdro-2H-pyran-4-vn-7H- Pyrrolor2,3-dlpyrimidin-4-yll-phenyl)-2-fluoro-4-(1-hvdroxy-1-methyl-ethyl)-benzam^
Intermediate 14
Figure imgf000057_0001
Intermediate 14 was prepared analogue to Intermediate 9 by replacing Intermediate 7 with Intermediate 13.
(3) N-(3-r6-(3.6-Dihvdro-2H-pyran-4-vn-7H-pyrrolor2.3-dlpyrimidin-4-yl1-2- hvdroxymethyl-phenyl)-2-fluoro-4-(1-hvdroxy-1-methyl-ethyl)-benzamide
Figure imgf000057_0002
Example 23 was prepared analogue to Example 16 step 4 by replacing Intermediate 9 with Intermediate 14.
MS (ESI): 503 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.42 (s, 1 H), 10.39 (d, 1 H), 8.82 (s, 1 H), 8.19 (d, 1 H), 7.86 (t, 1 H), 7.52-7.30 (m, 4H), 6.62 (br s, 1 H), 6.42 (s, 1 H), 5.65 (m, 1 H), 5.26 (s, 1 H), 4.63 (m, 2H), 4.27 (m, 2H), 3.80 (m, 2H), 2.41 (m, 2H), 1.45 (s, 6H). Example 24
4-tert-Butyl-N-(3-r6-(3,6-dihvdro-2H-thiopyran-4-yl)-7H-pyrrolor2,3-dlpyrim
fluoro-2-methyl-phenyl)-benzamide
Figure imgf000058_0001
(1 ) 4-Chloro-6-(3.6-dihvdro-2H-thiopyran-4-vn-7H-pyrrolor2.3-dlpyrimidine.
Intermediate 15
Figure imgf000058_0002
Intermediate 15 was prepared analogue to Intermediate 1 by replacing 4-(4, 4,5,5- tetramethyl-[1 ,2,3]dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester with 2-(3,6-dihydro-2H-thiopyran-4-yl)-4,4,5,5-tetramethyl-[1 ,3,2]dioxaborolane. MS (ESI): 252 [M+H]+
(2) 3-r6-(3,6-Dihvdro-2H-thiopyran-4-vn-7H-pyrrolor2,3-dlpyrimidin-4-yll-5-fluoro-2- methyl-phenylamine, Intermediate 16
Figure imgf000058_0003
Intermediate 16 was prepared analogue to Intermediate 6 by replacing Intermediate 3 with Intermediate 15.
MS (ESI): 341 [M+H]+
(3) 4-tert-Butyl-N-(3-r6-(3.6-dihvdro-2H-thioDyran-4vn-7H-Dyrrolor2.3-dlDyrimidin-4- vH-5-fluor-2-methyl-phenyl)-benzamide
Figure imgf000059_0001
Example 24 was prepared analogue to Example 15 by replacing Intermediate 6 with Intermediate 16 and 4-dimethyaminobenzoyl chloride with tert-butylbenzoyl chloride. MS (ESI): 501 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.39 (s, 1 H), 9.97 (s, 1 H), 8.82 (s, 1 H), 7.95 (d, 2H), 7.56 (d, 2H), 7.47 (m, 1 H), 71.19 (m, 1 H), 6.78 (m, 1 H), 6.34 (s, 1 H), 3.37 (m, 2H), 2.82 (m, 2H), 2.66 (m, 2H), 2.1 1 (s, 3H), 1.32 (s, 9H).
Example 25
4-tert-Butyl-N-(5-fluoro-2-methyl-3-r6-(3.6-dihvdro-1-oxido-2H-thiopyran-4-vn-7H-
Pyrrolor2,3-dlpyrimidin-4-yll-phenyl)-benzamide
Figure imgf000059_0002
Example 24 (160 mg, 0.32 mmol) was dissolved in acetic acid (5 ml) and hydrogen peroxide (0.033 ml, 0.32 mmol) was added. The mixture was stirred at room temperature for 2 hours. The mixture was treated with sodium hydrogen sulfite solution (10%, 10 ml) for 10 minutes, diluted with water, basified with 2N sodium hydroxide solution and extracted with EtOAc. The organic layer was washed with brine (2x), dried over sodium sulfate, filtered and evaporated. The residue was purified by flash chromatography on silica (EtOAc/MeOH/N H4OH 9: 1 :0.1 to EtOAc/MeOH/NH4OH 85: 15: 1.5) to afford the compound Example 25 as a beige solid.
MS (ESI): 517 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.49 (br s, 1 H), 9.99 (s, 1 H), 8.85 (s, 1 H), 7.96 (d, 2H), 7.55 (d, 2H), 7.49 (m, 1 H), 7.21 (m, 1 H), 6.49 (br s, 1 H), 6.46 (s, 1 H), 3.69 (m, 1 H), 3.50 (m, 1 H), 3.13 (m, 1 H), 295 (m, 1 H), 2.89 (m, 1 H), 2.80 (m, 1 H), 2.13 (s, 3H), 1.31 (s, 9H).
Example 26
4-tert-Butyl-N-(3-r6-(3.6-dihvdro-1.1-dioxido-2H-thioDyran-4-vn-7H-Dyrrolor2.3- dlpyrimidin-4-yll-5-fluoro-2-methyl-phenyl)-benzamide
Figure imgf000060_0001
Example 24 (160 mg, 0.32 mmol) was dissolved in DCM (15 ml), then trifluoro acetic acid (5 ml) and hydrogen peroxide (0.065 ml, 0.64 mmol) were added. The mixture was stirred at room temperature for 2 hours. The mixture was treated with sodium hydrogen sulfite solution (10%, 10 ml) for 10 minutes, diluted with water, basified with 2N sodium hydroxide solution and extracted with EtOAc. The organic layer was washed with brine (2x), dried over sodium sulfate, filtered and evaporated. The residue was purified by flash chromatography on silica (EtOAc/MeOH/NH4OH 98:2:0.2 to EtOAc/MeOH/N H OH 95:5:0.5) to afford the compound Example 26 as a beige solid.
MS (ESI): 533 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.45 (br s, 1 H), 9.93 (s, 1 H), 8.84 (s, 1 H), 7.91 (d, 2H), 7.56 (d, 2H), 7.49 (m, 1 H), 7.21 (m, 1 H), 6.41 (m, 2H), 4.00 (m, 2H), 3.36 (m, 2H), 3.07 (m, 2H), 2.16 (s, 3H), 1.33 (s, 9H). Example 27
4-tert-Butyl-N-(3-r6-(1 ^-dioxa-spiror4.5ldec-7-en-8-vn-7H-pyrrolor2,3-dlpyrimid fluoro-2-methyl-phenyl)-benzamide
Figure imgf000061_0001
(1 ) 4-Chloro-6-(1 ,4-dioxa-spiror4.51dec-7-en-8-yl)-7H-pyrrolor2,3-dlpyrimidine,
Intermediate 17
Figure imgf000061_0002
Intermediate 17 was prepared analogue to Intermediate 1 by replacing 4-(4, 4,5,5- tetramethyl-[1 ,2,3]dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester with 1 ,4-dioxaspiro[5,5]dec-7-en-8-boronic acid pinacol ester.
MS (ESI): 292 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.59 (br s, 1 H), 8.54 (s, 1 H), 6.54 (s, 1 H), 6.50 (br s, 1 H), 3.93 (m, 4H), 2.61 (m, 2H), 2.43 (m, 2H), 1.83 (m, 2H).
(2) 3-r6-(1.4-Dioxa-spiror4.51dec-7-en-8-vn-7H-pyrrolor2.3-dlpyrimidin-4-yl1-5-fluoro-
2-methyl-phenylamine, Intermediate 18
Figure imgf000061_0003
Intermediate 18 was prepared analogue to Intermediate 6 by replacing Intermediate 3 with Intermediate 17.
MS (ESI): 381 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.24 (br s, 1 H), 8.75 (s, 1 H), 6.55 (m, 1 H), 6.46 (br s, 1 H), 6.38 (m, 1 H), 6.21 (s, 1 H), 5.36 (s, 2 NH), 3.91 (m, 4H), 2.55 (m, 2H), 2.41 (m, 2H), 1.88 (s, 3H), 1.78 (m, 2H).
(3) 4-tert-Butyl-N-(3-r6-(1.4-dioxa-spiror4.51dec-7-en-8-vn-7H-pyrrolor2.3-dlpyrimidin-
4-vH-5-fluoro-2-methyl-phenyl)-benzamide
Figure imgf000062_0001
Example 27 was prepared analogue to Example 15 by replacing Intermediate 6 with Intermediate 18 and 4-dimethyaminobenzoyl chloride with 4-tert-butylbenzoyl chloride. MS (ESI): 541 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.36 (br s, 1 H), 9.98 (s, 1 H), 8.82 (s, 1 H), 7.96 (d, 2H), 7.58 (d, 2H), 7.48 (m, 1 H), 7.21 (m, 1 H), 6.50 (br s, 1 H), 6.29 (s, 1 H), 3.93 (m, 4H), 2.58 (m, 2H), 2.43 (m, 2H), 2.12 (s, 3H), 1.81 (m, 2H), 1.33 (s, 9H).
Example 28
4-tert-Butyl-N-{5-fluoro-3-r6-(4-hv^^
2-methyl-phenvD-benzamide
Figure imgf000063_0001
(1 ) 4-tert-Butyl-N-(5-fluoro-3-r6-(4-hvdroxy-cvclohex-1-envn-7H-pyrrolor2.3- dlpyrimidin-4-yll-2-methyl-phenyl)-benzamide, Intermediate 19
Figure imgf000063_0002
A mixture of Example 27 (450 mg, 0.832 mmol) and TFA (6 ml) in DCM (30ml) was stirred at r.t. for 6 hrs. The solvents were removed in vacuo and the crude product was purified by flash chromatography (silica gel, EtOAc/MeOH/NH4OH gradient) to yield Intermediate 19.
MS (ESI): 497 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.47 (br s, 1 H), 9.97 (s, 1 H), 8.83 (s, 1 H), 7.94 (d, 2H), 7.57 (d, 2H), 7.48 (m, 1 H), 7.20 (m, 1 H), 6.65 (br s, 1 H), 6.40 (s, 1 H), 3.1 1 (m, 2H), 2.86 (m, 2H), 2.53 (m, 2H), 2.1 1 (s, 3H), 1.33 (s, 9H). (2) 4-tert-Butyl-N-(5-fluoro-3-r6-(4-hvdroxy-cvclohex-1-envn-7H-pyrrolor2.3- dlpyrimidin-4-yll-2-methyl-phenyl)-benzamide
Figure imgf000064_0001
To a solution of Intermediate 19 (100 mg, 0.201 mmol) in MeOH (25 ml) was added NaBH4 (9.1 mg, 0.242 mg). The mixture was stirred at r.t. for 2 hrs, then the solvent was removed in vacuo, and the crude mixture was purified by flash chromatography (silica gel, EtOAc/MeOH/NH4OH gradient) to yield Example 28.
MS (ESI): 499 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.31 (br s, 1 H), 9.96 (s, 1 H), 8.79 (s, 1 H), 7.94 (d, 2H), 7.57 (d, 2H), 7.47 (m, 1 H), 7.18 (m, 1 H), 6.50 (br s, 1 H), 6.24 (s, 1 H), 4.73 (br s, 1 H), 3.80 (m, 1 H), 2.51 (m, 2H), 2.44 (m, 1 H), 2.39 (m, 1 H), 2.10 (s, 3H), 1.84 (m, 1 H), 1.59 (m, 1 H), 1.33 (s, 9H).
Example 29
4-f4-r3-(4-Cvclopropyl-benzoylamino)-5-fluoro-2-methyl-phenyll-7H-pyrrolor2'3- dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000064_0002
(1 ) 4-r4-(3-Amino-5-fluoro-2-methyl-phenvn-7H-pyrrolor2.3-dlpyrimidin-6-yl1-3.6- dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester, Intermediate 20
Figure imgf000065_0001
For the Suzuki coupling of chloro compound Intermediate 1 (0.63 g, 1.88 mmol) and boronic ester derivative Intermediate 5 (0.52 g, 2.07 mmol) the same procedure was used as described in Example 1 step 6, to afford the compound Intermediate 20 as a beige solid.
MS (ESI): 424 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.33 (br s, 1 H), 8.76 (s, 1 H), 6.54 (m, 2H), 6.40 (d, 1 H), 6.26 (s, 1 H), 5.36 (s, 2NH), 4.05 (br s, 2H), 3.51 (m, 2H), 1 .99 (m, 2H), 1.88 (s, 3H), 1.42 (s, 9H).
(2) 4-f4-r3-(Cvclopropyl-benzoylamino)-5-fluoro-2-methyl-phenyll-7H-pyrrolor2,3- dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid tert-butyl ester, Intermediate 21
Figure imgf000065_0002
To a solution of 4-cyclopropyl benzoic acid (0.58 g, 3.57 mmol) in toluene (2 ml), thionyl chloride (1.29 ml, 17.87 mmol) was added. The mixture was stirred at 80°C for 2 hours and afterwards evaporated under reduced pressure. The residue was dissolved together with Intermediate 20 (1.0 g, 2.36 mmol) in pyridine (5 ml) and the mixture was stirred at room temperature for 2 hours. Pyridine was evaporated under reduced pressure and the residue was dissolved in DCM and washed with saturated sodium hydrogen carbonate solution and brine (2x). The residue was purified by flash chromatography on silica (cyclohexane to EtOAc) to afford the compound Intermediate 21 as a beige solid.
MS (ESI): 568 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.43 (br s, 1 H), 9.93 (s, 1 NH) 8.82 (s, 1 H), 7.89 (d, 2H), 7.46 (m, 1 H), 7.24 (d, 2H), 7.18 (m, 1 H), 6.57 (br s, 1 H), 6.33 (s, 1 H), 4.06 (br s, 2H), 3.52 (m, 2H), 2.48 (m, 2H), 2.10 (s, 3H), 2.01 (m, 1 H), 1.42 (s, 9H), 1.02 (m, 2H), 0.76 (m, 2H).
(3) 4-Cvclopropyl-N-(5-fluoro-2-methyl-3-r6-(1.2.3.6-tetrahvdro-pyridin-4-vn-7H-
Pyrrolor2,3-dlpyrimidin-4-yll-phenyl)-benzamide, Intermediate 22
Figure imgf000066_0001
The BOC protecting group of compound Intermediate 21 (1.0 g, 1.76 mmol) was removed as described in Example 1 step 2 to afford the compound Intermediate 22 as a beige solid.
MS (ESI): 468 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.33 (br s, 1 H), 9.94 (s, 1 NH) 8.80 (s, 1 H), 7.89 (d, 2H), 7.47 (m, 1 H), 7.24 (d, 2H), 7.19 (m, 1 H), 6.62 (br s, 1 H), 6.24 (s, 1 H), 3.41 (br s, 2H), 2.88 (m, 2H), 2.33 (m, 2H), 2.10 (s, 3H), 2.01 (m, 1 H), 1.04 (m, 2H), 0.77 (m, 2H).
(4) 4-(4-r3-(4-Cvclopropyl-benzoylamino)-5-fluoro-2-methyl-phenyll-7H-pyrrolor2'3- dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000067_0001
The amino compound Intermediate 22 (0.2 g, 0.51 mmol) was acylated as described in Example 1 step 3 to afford the compound Example 29 as a beige solid.
MS (ESI): 539 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.42 (br s, 1 H), 9.93 (s, 1 NH) 8.82 (s, 1 H), 7.89 (d, 2H), 7.46 (m, 1 H), 7.24 (d, 2H), 7.18 (m, 1 H), 6.59 (br s, 1 H), 6.32 (s, 1 H), 3.90 (br s, 2H), 3.33 (m, 2H), 2.76 (s, 6H), 2.53 (m, 2H), 2.10 (s, 3H), 2.02 (m, 1 H), 1.03 (m, 2H), 0.77 (m, 2H).
Example 30
4-Cvclopropyl-N-(5-fluoro-2-methyl-3-f6-ri-(Pyrrolidine-1-carbonyl)-1 ,2,3,6-tetrahydro-
Pyridin-4-yll-7H-pyrrolor2,3-dlpyrimidin-4-yl)-phenyl)-benzamide
Figure imgf000067_0002
The amino compound Intermediate 22 (0.2 g, 0.51 mmol) was acylated using pyrrolidine-1-carbonyl chloride as described in Example 1 step 3 to afford the compound Example 30 as a beige solid. MS (ESI): 565 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.41 (br s, 1 H), 9.93 (s, 1 NH) 8.82 (s, 1 H), 7.89 (d, 2H), 7.45 (m, 1 H), 7.24 (d, 2H), 7.18 (m, 1 H), 6.58 (br s, 1 H), 6.32 (s, 1 H), 3.93 (br s, 2H), 3.38 (m, 2H), 3.29 (m, 4H), 2.52 (m, 2H), 2.10 (s, 3H), 2.0 (m, 1 H), 1.75 (m, 4H), 1.04 (m, 2H), 0.76 (m, 2H).
Example 31
Acetic acid 2-(4-(4-r3-(4-cvclopropyl-benzoylamino)-5-fluoro-2-methyl-phenyll-7H-
Pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridin-1-yl)-2-oxo-ethyl ester
Figure imgf000068_0001
The amino compound Intermediate 22 (0.15 g, 0.32 mmol) was acylated using acetic acid chlorocarbonylmethyl ester as described in Example 1 step 3 to afford the compound Example 31 as a beige solid.
MS (ESI): 568 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.45 (br s, 1 H), 9.94 (s, 1 NH) 8.83 (s, 1 H), 7.89 (d, 2H), 7.47 (m, 1 H), 7.22 (d, 2H), 7.19 (m, 1 H), 6.58 (br s, 1 H), 6.34 (s, 1 H), 4.86 (m, 2H), 4.15 (m, 2H), 3.57 (m, 2H), 2.59 (m, 2H), 2.10 (s, 3H), 2.08 (s, 3H), 2.01 (m, 1 H), 1.03 (m, 2H), 0.77 (m, 2H).
Example 32
4-Cvclopropyl-N-(5-fluoro-3-(6-ri-(2-hvdroxy-actyl)-1 ,2,3,6-tetrahvdro-pyridin-4-yll-7H- Pyrrolor2,3-dlpyrimidin-4-yl)-2-methyl-phenyl)benzamide
Figure imgf000069_0001
The acetyl compound of Example 31 (100 mg, 0.17 mmol) was dissolved in THF (1 ml) and EtOH (1 ml) and 2N sodium hydroxide solution (0.31 ml, 0.62 mmol) was added. The mixture was stirred at room temperature for 5 minutes. The mixture was diluted with water and extracted with DCM (3x). The organic layer was washed with brine (2x), dried over sodium sulfate, filtered and evaporated. The residue was purified by flash chromatography on silica (EtOAc to EtOAc/MeOH/NH4OH 98:2:0.2) to afford the compound Example 32 as a beige solid.
MS (ESI): 526 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.47 (br s, 1 H), 9.95 (s, 1 NH) 8.82 (s, 1 H), 7.89 (d, 2H), 7.47 (m, 1 H), 7.24 (d, 2H), 7.18 (m, 1 H), 6.61 (m, 1 H), 6.33 (m, 1 H), 4.63 (m, 2H), 4.04 (m, 4H), 3.68 (m, 1 H), 3.54 (m, 1 H), 2.57 (m, 2H), 2.10 (s, 3H), 2.01 (m, 1 H), 1.04 (m, 2H), 0.77 (m, 2H).
Example 33
N-(3-(6-M-(2-Cvano-acetyl)-1 ,2,3,6-tetra^
yl)-5-fluoro-2-methyl-phenyl)-4-cyclopropyl-benzamide
Figure imgf000070_0001
Amino compound Intermediate 22 (30 mg, 0.064 mmol), cyanoacetic acid (1 1 mg, 0.128 mmol) and HATU (61 mg, 0.16 mmol) were dissolved in DIPEA (0.056 ml, 0.321 mmol) and DMF (1.0 ml). The solution was stirred at room temperature for 24 hrs. The reaction was diluted with DCM and washed with saturated sodium hydrogen carbonate solution and brine (2x). The organic layer was dried with sodium sulfate and evaporated. The residue was purified by flash chromatography on silica (cyclohexane to EtOAc) to afford the compound Example 33 as a beige solid.
MS (ESI): 536 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 9.56 (br s, 1 NH) 8.76 (s, 1 H), 7.85 (d, 2H), 7.43 (m, 1 H), 7.19 (d, 2H), 7.09 (m, 1 H), 6.53 (m, 1 H), 6.29 (m, 1 H), 4.16 (m, 2H), 3.94 (m, 2H), 3.63 (m, 2H), 2.56 (m, 2H), 2.10 (s, 3H), 2.01 (m, 1 H), 1.01 (m, 2H), 0.74 (m, 2H).
Example 34
N-(5-Fluoro-2-methyl-3-(6-ri-(Pyrrolidine-1-carbonyl)-1 ,2,3,6-tetrahydro-pyridin-4-vn Pyrrolor2,3-dlpyrimidin-4-yl)-phenyl)-4-(pentafluoro-sulfanyl)-benzamide
Figure imgf000071_0001
(1 ) 4-(4-(5-Fluoro-2-methyl-3-r4-(pentafluoro-sulfanyl)-benzoylaminol-phenyl)-7H- Pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid tert-butyl ester, Intermediate 23
Figure imgf000071_0002
Intermediate 23 was prepared analogue to Intermediate 9 by replacing Intermediate 7 with Intermediate 20 and Intermediate 8 with 4-(pentafluoro-sulfanyl)-benzoic acid. MS (ESI): 654 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.44 (br s, 1 H), 10.33 (s, 1 H), 8.83 (s, 1 H), 8.18 (d, 2H), 8.12 (d, 2H), 7.50 (m, 1 H), 7.24 (m, 1 H), 6.57 (m, 1 H), 6.34 (s, 1 H), 4.06 (m, 2H), 3.51 (m, 2H), 2.49 (m, 2H), 2.12 (s, 3H), 1.42 (s, 9H). (2) N-(5-Fluoro-2-methyl-3-r6-(1.2.3.6-tetrahvdro-pyridin-4-vn-7H-pyrrolor2.3- dlpyrimidin-4-yll-phenyl)-4-(pentafluoro-sulfanyl)-benzamide, Intermediate 24
Figure imgf000072_0001
Intermediate 24 was prepared analogue to Intermediate 22 by replacing Intermediate 21 with Intermediate 23.
MS (ESI): 554 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.42 (br s, 1 H), 10.43 (s, 1 H), 8.83 (s, 1 H), 8.23 (d, 2H), 8.13 (d, 2H), 7.51 (m, 1 H), 7.24 (m, 1 H), 6.54 (m, 1 H), 6.31 (s, 1 H), 3.52 (m, 2H), 2.99 (m, 2H), 2.45 (m, 2H), 2.13 (s, 3H).
(3) N-(5-Fluoro-2-methyl-3-f6-ri-(Pyrrolidine-1-carbonyl)-1 ,2,3,6-tetrahydro-pyridin-4- yll-7H-pyrrolor2,3-dlpyrimidin-4-yl)-phenyl)-4-(pentafluoro-sulfanyl)-benzamide
Figure imgf000072_0002
Example 34 was prepared analogue to Example 30 by replacing Intermediate 22 with Intermediate 24.
MS (ESI): 651 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.42 (br s, 1 H), 10.32 (s, 1 H), 8.82 (s, 1 H), 8.18 (d, 2H), 8.12 (d, 2H), 7.48 (m, 1 H), 7.23 (m, 1 H), 6.59 (m, 1 H), 6.33 (s, 1 H), 3.93 (m, 2H), 3.38 (m, 2H), 3.29 (m, 4H), 2.50 (m, 2H), 2.12 (s, 3H), 1.75 (m, 4H). Example 35
Acetic acid 2-r4-(4-{5-fluoro-2-methyl-3-r4-(pentafluoro-sulfanyl)-benzo
7H-pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridin-1-yll-2-oxo-ethyl ester
Figure imgf000073_0001
Example 35 was prepared analogue to Example 31 by replacing Intermediate 22 with Intermediate 24.
MS (ESI): 654 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.47 (br s, 1 H), 10.34 (s, 1 H), 8.83 (s, 1 H), 8.19 (d, 2H), 8.12 (d, 2H), 7.49 (m, 1 H), 7.25 (m, 1 H), 6.60 (m, 1 H), 6.37 (s, 1 H), 4.86 (s, 2H), 4.16 (m, 2H), 3.58 (m, 2H), 2.60 (m, 2H), 2.12 (s, 3H), 2.08 (s, 3H).
Example 36
N-(5-Fluoro-3-(6-ri-f2-hvdroxy-acetvn-1.2.3.6-tetrahvdro-pyridin-4-yl1-7H-pyrrolor2.3- dlpyrimidin-4-yl)-2-methyl-phenyl)-4-(pentafluoro-sulfanyl)-benzamide
Figure imgf000073_0002
Example 36 was prepared analogue to Example 32 by replacing Example 31 with Example 35.
MS (ESI): 612 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.43 (br s, 1 H), 10.47 (s, 1 H), 8.84 (s, 1 H), 8.22 (d, 2H), 8.12 (d, 2H), 7.50 (m, 1 H), 7.24 (m, 1 H), 6.60 (m, 1 H), 6.35 (s, 1 H), 4.86 (s, 2H), 4.20 (m, 2H), 4.16 (m, 2H), 3.51 (m, 2H), 2.58 (m, 2H), 2.13 (s, 3H), 2.08 (s, 3H).
Example 37
4-(4-r3-(4-tert-Butyl-benzoylamino)-2-(tert-butyl-diphenyl-silanyloxymethyl)-phenyll-7H- Pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid tert-butyl ester
Figure imgf000074_0001
(1 ) (2-Bromo-nitro-benzyloxyHert-butyl-diphenyl-silane, Intermediate 25
Figure imgf000074_0002
To a solution of (2-bromo-6-nitro-phenyl)-methanol (10.0 g, 43.1 mmol) and imidazole (5.87 g, 86.0 mmol) in DMF (20 ml), tert-butyl-chloro-diphenyl-silane was added dropwise at room temperature. The mixture was stirred at 50°C for 1 hour. The mixture was diluted with EtOAc and washed with brine (2x). The organic layer was dried with sodium sulfate and evaporated. The residue was taken up in heptane and stirred at room temperature for 3 hours. The solid was filtered off to afford compound Intermediate 25 as a beige solid.
MS (ESI): 471 [M+H]+ , 1H-NMR (CDCI3): δ (ppm) 7.80 (m, 6H), 7.40 (m, 6H), 7.24 (m, 1 H), 5.10 (s, 2H), 0.99 (s, 9H).
(2) 2-r2-(tert-Butyl-diphenyl-silanyloxymethyl)-3-nitro-phenyll-4,4,5,5-tetramethyl- ri ,3,21dioxaborolane, Intermediate 26
Figure imgf000075_0001
To a mixture of Intermediate 25 (5.0 g, 10.63 mmol) and bis(diphenylphosphino)ferrocenedichloropalladium (II) (0.43 g, 0.52 mmol) in 60 ml dioxane, bis-(pinacolato)-diboron (5.4 g, 21.26 mmol) and potassium acetate (6.25 g, 63.7 mmol) were added. The mixture was heated to 90°C for 4 hours. After cooling the brownish mixture was filtered over Kieselgur (Supelco) and evaporated. The residue was purified by flash chromatography on silica (heptane to heptane/EtOAc 4: 1 ) to afford the compound Intermediate 26 as a yellow oil.
MS (ESI): no peaks , 1H-NMR (CDCI3): δ (ppm) 7.65 (m, 5H), 7.30 (m, 8H), 5.30 (s, 2H), 1.10 (s, 12H), 0.97 (s, 9H).
(3) 2-(tert-Butyl-diphenyl-silanyloxymethyl)-3-(4,4,5,5-tetramethyl-ri ,3,21dioxborolan- 2-yl)-phenylamine, Intermediate 27
Figure imgf000075_0002
The nitro compound Intermediate 26 (10.0 g, 19.32 mmol) was dissolved in 300 ml EtOAc and Pd/C 10% (Pd) (2.0 g) were added. The mixture was hydrogenated at room temperature and normal pressure for 20 hours. The mixture was filtered over Kieselgur (Supelco) and evaporated. The residue was purified by flash chromatography on silica (cyclohexane to cyclohexane/EtOAc 4:1 ) to afford the compound Intermediate 27 as a yellow oil.
MS (ESI): 488 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 7.63 (m, 4H), 7.44 (m, 2H), 7.39 (m, 4H), 7.03 (t, 1 H), 6.91 (d, 1 H), 6.84 (d, 1 H), 5.10 (s, 2H), 5.08 (s, 2H), 0.99 (s, 12H), 0.98 (s, 9H).
(4) 4-tert-Butyl-N-r2-tert-butyl-diphenyl-silanyloxymethyl)-3-(4,4,5,5-tetramethyl- n ,3,21dioxaborolan-2-yl)-phenyll-benzamide, Intermediate 28
Figure imgf000076_0001
The anilino compound Intermediate 27 (92.0 g, 189 mmol) was dissolved in DCM (1.2 I) and triethylamine (80 ml). To the mixture 4-tert-butylbenzoylchloride was added over 5 minutes at 0°C. The mixture was stirred at room temperature for 1 hour. The mixture was washed with saturated ammonium chloride solution and brine (2x) and evaporated. The residue was taken up in hexane and stirred at room temperature for 3 hours. The solid was filtered off to afford compound Intermediate 28 as a beige solid.
MS (ESI): 648 [M+H]+ , 1H-NMR (CDCI3): δ (ppm) 9.85 (br s, 1 H), 7.80 (m, 2H), 7.60 (m, 6H), 7.35-7.25 (m, 9H), 5.30 (s, 2H), 1 .35 (s, 9H), 1.05 (s, 18H). (5) 4-(4-r3-(4-tert-Butyl-benzoylamino)-2-(tei1-butyl-diphenyl-silanyloxymethyl)- phenyll-7H-pyrrolor2,3-dlpyrimid-6-yl)-3,6-dihvdro-2H-pyridin3-1-carboxylic acid tert- butyl ester, Intermediate 29
Figure imgf000077_0001
For the Suzuki coupling between chloro compound Intermediate 1 (0.50 g, 1.49 mmol) and boronic ester derivative Intermediate 28 (1.45 g, 2.24 mmol) the same procedure was used as described in Example 1 step 6, to afford the compound Intermediate 29 as a beige solid.
MS (ESI): 820 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.33 (s, 1 H), 10.02 (s, 1 H), 8.68 (s, 1 H), 7.78 (m, 2H), 7.75 (m, 1 H), 7.46 (m, 2H), 7.40 (m, 1 H), 7.25 (m, 2H), 7.12 (m, 1 H), 7.1 1 (m, 8H), 6.54 (br s, 1 H), 6.30 (s, 1 H), 4.97 (s, 2H), 4.07 (m, 2H), 3.52 (m, 2H), 2.43 (m, 2H), 1.41 (s, 9H), 1.30 (s, 9H), 0.53 (s, 9H).
(6) 4-tert-Butyl-N-(2-(tert-butyl-diphenyl-silanyloxymethvn-3-r6-(1.2.3.6-tetrahvdro-
Pyridin-4-yl)-7H-pyrrolor2,3-dlpyrimidin-4-yll-benzamide, Intermediate 30
Figure imgf000077_0002
The BOC protecting group of compound Intermediate 29 (0.92 g, 1.12 mmol) was removed as described in Example 1 step 2 to afford the compound Intermediate 30 as a beige solid.
MS (ESI): 720 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.50 (s, 1 H), 10.08 (s, 1 H), 8.72 (s, 1 H), 7.80 (m, 2H), 7.75 (m, 1 H), 7.53 (m, 1 H), 7.42 (m, 1 H), 7.26 (m, 2H), 7.15 (m, 2H), 7.1 1 (m, 8H), 6.56 (br s, 1 H), 6.42 (s, 1 H), 4.99 (s, 2H), 3.85 (m, 2H), 3.32 (m, 2H), 2.67 (m, 2H), 1.29 (s, 9H), 0.52 (s, 9H).
(7) 4-{4-r3-(4-tert-Butyl-benzoylamino)-2-(tert-butyl-diphenyl-silanyloxymethyl)- phenyll-7H-pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide, Intermediate 31
Figure imgf000078_0001
The amino compound Intermediate 30 (0.1 g, 0.14 mmol) was acylated as described in Example 1 step 3 to afford the compound Intermediate 31 as a beige solid.
MS (ESI): 791 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.35 (s, 1 H), 10.04 (s, 1 H), 8.70 (s, 1 H), 7.79 (m, 2H), 7.74 (m, 1 H), 7.53 (m, 1 H), 7.47 (m, 2H), 7.43 (m, 1 H), 7.27 (m, 2H), 7.14-7.11 (m, 8H), 6.57 (br s, 1 H), 6.30 (s, 1 H), 4.98 (s, 2H), 3.91 (m, 2H), 3.32 (m, 2H), 2.76 (s, 6H), 2.52 (m, 2H), 1.31 (s, 9H), 0.53 (s, 9H). (8) 4-(4-r3-(4-tert-Butyl-benzoylamino)-2-hvdroxymethyl-phenyll-7H-pyrrolor2,3- dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000079_0001
The compound Intermediate 31 (65 mg, 0.082 mmol) was dissolved in a 1 M solution of tetrabutyl ammonium fluoride in THF (2 ml, 2.0 mmol). The mixture was stirred at room temperature for 20 hours. The solvent was removed under reduced pressure. The residue was purified by flash chromatography on silica (EtOAc/MeOH/NH4OH 95:5:0.5) to afford the compound Example 37 as a beige solid.
MS (ESI): 553 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.44 (s, 1 H), 10.53 (s, 1 NH) 8.83 (s, 1 H), 8.23 (m, 1 H), 7.91 (d, 2H), 7.60 (d, 2H), 7.51 (m, 1 H), 7.35 (m, 1 H), 6.60 (br s, 1 H), 6.45 (s, 1 H), 4.69 (s, 2H), 3.90 (br s, 2H), 3.33 (m, 2H), 2.77 (s, 6H), 2.54 (m, 2H), 1.33 (s, 9H).
Example 38
4-tert-Butyl-N-(3-(6-ri-(2-fluoro-acetvn-1.2.3.6-tetrahvdro-pyridin-4-yll-7H-pyrrolor2.3- dlpyrimidin-4-yl)-2-hvdroxymethyl-phenyl)-benzamide
Figure imgf000079_0002
(1 ) 4-tert-Butyl-N-(2-(tert-butyl-diphenyl-silanyloxymethvn-3-(6-ri-f2-fluoro-acetvn- 1 ,2,3,6-tetrahvdro-pyridin-4-yll-7H-pyrrolor2,3-dlpyrimidin-4-yl)-phen
Intermediate 32
Figure imgf000080_0001
To a solution of Intermediate 30 (100 mg, 0.139 mmol) in pyridine (5 ml) was added fluoro-acetyl chloride (20 mg, 0.208 mmol). The mixture was stirred at r.t. for 20 hrs, then the solvent was removed in vacuo. The crude product was purified using flash chromatography (siliga gel, EtOAc/MeOH/NH3 gradient) to obtain Intermediate 32.
(2) 4-tert-Butyl-N-(3-(6-ri-(2-fluoro-acetvn-1.2.3.6-tetrahvdro-pyridin-4-yll-7H-
Pyrrolor2,3-dlpyrimidin-4-yl)-2-hvdroxymethyl-phenyl)-benzamide
Figure imgf000080_0002
Example 38 was prepared analogue to Example 37 step 8 by replacing Intermediate 31 with Intermediate 32.
MS (ESI): 542 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.46 (br s, 1 H), 10.52 (s, 1 H), 8.83 (s, 1 H), 8.21 (d, 1 H), 7.90 (d, 2H), 7.59 (d, 2H), 7.50 (t, 1 H), 7.34 (d, 1 H), 6.61 (br, s), 6.44 (s, 1 H), 5.91 (br s, 1 H), 5.25 (m, 2H), 4.67 (s, 2H), 4.18 (m, 1 H), 4.08 (m, 1 H), 3.67 (m, 1 H), (3.47 m, 1 H), 2.57 (m, 1 H), 2.51 (m, 1 H), 1.32 (s, 9H).
Example 39
4-(4-(5-Fluoro-3-r2-fluoro-4-(1-hvdroxy-1 -methyl-ethyl )-benzoylaminol-2-methyl-phenyl)- 7H-pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000081_0001
(1 ) 4-Chlorocarbonyl-3-fluoro-benzoic acid methyl ester, Intermediate 33
Figure imgf000081_0002
2-Fluoro-terephthalic acid 4-methyl ester (J. Med. Chem., 52(19), 5950-5966; 2009) (500 mg, 2.25 mmol) was suspended in DCM (10 ml). Then oxalyl chloride (5.08 ml, 58.0 mmol) and a drop of DMF were added. The resulting mixture was refluxed for 14 hrs. The solvents were removed in vacuo, and the resulting crude Intermediate 33 was used in the next step without purification. (2) N-(3-Bromo-5-fluoro-2-methyl-phenyl)-3-fluoro-terephthalamic acid methyl ester, Intermediate 34
Figure imgf000082_0001
3-Bromo-5-fluoro-2-methylaniline (515 mg, 2.53 mmol) was dissolved in pyridine (20 ml). Then Intermediate 33 (547 mg, 2.53 mmol) and DMAP (3.09 mg, 0.025 mmol) were added and the resulting mixture was stirred at r.t. for 2 hrs. The solvents were removed in vacuo and the residue was taken up with EtOAc and H20. The organic layer was washed with sat. NaHC03 solution and brine, and then dried over Na2S04. The solvents were removed in vacuo and the resulting residue was recrystallized with MeOH to afford Intermediate 34 as colorless solid.
(3) N-(3-Bromo-5-fluoro-2-methyl-phenyl)-2-fluoro-4-(1-hvdroxy-1-methyl-ethyl)- benzamide, Intermediate 35
Figure imgf000082_0002
Intermediate 34 (75 mg, 0.195 mmol) was dissolved in THF (10 ml) and the resulting mixture was cooled to 0°C. Then methyl magnesium bromide (0.390 ml, 1.17 mmol) was added dropwise and stirring was continued for 3 hrs. The mixture was concentrated to dryness and then diluted with MeOH/DMSO. The resulting precipitate was filterd off to give Intermediate 35 as a white solid.
MS (ESI): 384 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 10.04 (br s, 1 H),.7.63 (m, 1 H), 7.42 (m, 2H), 7.36 (m, 2H), 5.27 (s, 1 H), 2.24 (s, 3H), 1.40 (s, 6H). (4) 2-Fluoro-N-r5-fluoro-2-methyl-3-(4.4.5.5-tetramethyl-ri .3.21dioxaborolan-2-vn- phenyll-4-(1-hvdroxy-1-methyl-ethyl)-benzamide, Intermediate 36
Figure imgf000083_0001
Intermediate 36 was prepared analogue to Intermediate 4 by replacing 1-bromo-5- fluoro-2-methyl-3-nitro-benzene with Intermediate 35.
(5) 4-(4-f5-Fluoro-3-r2-fluoro-4-(1-hvdroxy-1-methyl-ethyl)-benzoylaminol-2-methyl- phenyl)-7H-pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000083_0002
Example 39 was prepared analogue to Example 37 by replacing the boronic ester Intermediate 28 in step 5 with the boronic ester Intermediate 36.
MS (ESI): 575 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.43 (s, 1 H), 9.93 (s, 1 H), 8.83 (s, 1 H), 7.74 (m, 1 H), 7.64 (d, 1 H), 7.43 (m, 2H), 7.18 (dd, 1 H), 6.60 (s, 1 H), 6.34 (s, 1 H), 5.30 (s, 1 H), 3.90 (m, 2H), 3.33 (m, 2H), 2.77 (s, 6H), 2.54 (m, 2H), 2.15 (s, 3H), 1.46 (s, 6H). Example 40
4-tert-Butyl-N-(5-fluoro-2-methyl-3-r6-(1 ,2,3,6-tetrahvdro-pyridin-4-yl)-7H-p
dlpyrimidin-4-yll-phenyl)-benzamide
Figure imgf000084_0001
(1 ) N-(3-Bromo-5-fluoro-2-methyl-phenyl)-4-tert-butyl-benzamide, Intermediate 37
Figure imgf000084_0002
Intermediate 37 was prepared analogue to Intermediate 34 by replacing Intermediate
33 with 4-tert-butyl-benzoyl chloride.
MS (ESI): 364 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 10.1 1 (br s, 1 H), 7.90 (d, 2H), 7.55 (d, 2H), 7.49 (d, 1 H), 7.32 (d, 1 H), 2.32 (s, 3H), 1.38 (s, 9H).
(2) 4-tert-Butyl-N-r5-fluoro-2-methyl-3-(4.4.5.5-tetramethyl-n .3.21dioxaborolan-2-vn- phenyll-benzamide, Intermediate 38
Figure imgf000084_0003
Intermediate 38 was prepared analogue to Intermediate 4 by replacing 1-bromo-5- fluoro-2-methyl-3-nitro-benzene with Intermediate 37.
MS (ESI): 412 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 9.85 (br s, 1 H), 7.90 (d, 2H), 7.55 (d, 2H), 7.35 (d, 1 H), 7.22 (d, 1 H), 2.35 (s, 3H), 1.40 (s, 6H), 1.32 (m, 15H).
(3) 4-tert-Butyl-N-(5-fluoro-2-methyl-3-r6-(1.2.3.6-tetrahvdro-Dyridin-4-vn-7H-
Pyrrolor2,3-dlpyrimidin-4-yll-phenyl)-benzamide
Figure imgf000085_0001
Example 40 was prepared analogue to Intermediate 30 in Example 37 step 6 by replacing Intermediate 28 in Example 37 step 5 with Intermediate 38.
MS (ESI): 484 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.55 (s, 1 H), 9.98 (s, 1 H), 8.85 (s, 1 H), 8.71 (br s, 1 H), 7.92 (d, 2H), 7.56 (d, 2H), 7.50 (m, 1 H), 7.18 (m, 1 H), 6.56 (m, 1 H), 6.43 (s, 1 H), 3.80 (m, 2H), 3.28 (m, 2H), 2.67 (m, 2H), 2.10 (s, 3H), 1.32 (s, 9H).
Example 41
4-f4-r3-(4-tert-Butyl-benzoylamino)-5-fluoro-2-methyl-phenyll-7H-pyrrolor2,3-dlpyrimidin-
6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000085_0002
Example 41 was prepared analogue to Example 37 step 7 by replacing Intermediate 30 with Example 40.
MS (ESI): 555 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.44 (s, 1 H), 9.98 (s, 1 H), 8.83 (s, 1 H), 7.95 (d, 2H), 7.58 (d, 2H), 7.48 (m, 1 H), 7.22 (m, 1 H), 6.60(s, 1 H), 6.33 (s, 1 H), 5.30 (s, 1 H), 3.90 (m, 2H), 3.32 (m, 2H), 2.77 (s, 6H), 2.55 (m, 2H), 2.12 (s, 3H), 1.34 (s, 9H).
Example 42
4-tert-Butyl-N-(5-fluoro-3-r6-(1-methanesulfonyl-1.2.3.6-tetrahvdro-Dyridin-4-vn-7H-
Pyrrolor2,3-dlpyrimidin-4-yll-2-methyl-phenyl)-benzamide
Figure imgf000086_0001
To a solution of Example 40 (90 mg, 0.186 mmol) and DIPEA (0.098 ml, 0.558 mmol) in THF (10 ml) was added methanesulfonyl chloride (0.015 ml, 0.186 mmol) dropwise. The resulting mixture was stirred at r.t. for 1 hr, then quenched with water and diluted with EtOAc. The organic layer was washed with sat. aqueous NaHC03 solution and brine, dried with Na2S04, and filtered. The solvents were removed in vacuo, and the crude product was purified by reversed phase HPLC (MeCN/H20 gradient) to yield Example 42 as a light yellow solid.
MS (ESI): 562 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.48 (br s, 1 H), 9.97 (s, 1 H), 8.82 (s, 1 H), 7.93 (d, 2H), 7.56 (d, 2H), 7.46 (m, 1 H), 7.19 (m, 1 H), 6.61 (s, 1 H), 6.36 (s, 1 H), 3.92 (m, 2H), 3.31 (m, 2H), 2.94 (s, 3H), 2.62 (m, 2H), 2.10 (s, 3H), 1.31 (s, 9H). Example 43
4-tert-Butyl-N-(3-r6-(1-dimethylsulfamoyl-1 ,2,3,6-tetrahvdro-pyridin-4-yl)-7H-pyrro dlpyrimidin-4-yll-5-fluoro-2-methyl-phenyl)-benzamide
Figure imgf000087_0001
Example 43 was prepared analogue to Example 42 by replacing methanesulfonyl chloride with Ν,Ν-dimethylamidosulfamoyl chloride.
MS (ESI): 591 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.48 (br s, 1 H), 9.97 (s, 1 H), 8.82 (s, 1 H), 7.93 (d, 2H), 7.56 (d, 2H), 7.46 (m, 1 H), 7.20 (m, 1 H), 6.59 (s, 1 H), 6.34 (s, 1 H), 3.93 (m, 2H), 3.40 (m, 2H), 2.76 (s, 6H), 2.57 (m, 2H), 2.09 (s, 3H), 1.31 (s, 9H).
Example 44
4-tert-Butyl-N-(3-r6-(1-methanesulfonyl-1.2.3.6-tetrahvdro-pyridin-4-vn-7H-pyrrolor2.3- dlpyrimidin-4-yll-2-methyl-phenyl)-benzamide
Figure imgf000087_0002
(1 ) 4-tert-Butyl-N-(2-methyl-3-r6-(1 ,2.3.6-tetrahvdro-pyridin-4-vn-7H-pyrrolor2.3- dlpyrimidin-4-yll-phenyl)-benzamide, Intermediate 39
Figure imgf000088_0001
Intermediate 39 was prepared analogue to Intermediate 30 in Example 37 step 6 by replacing Intermediate 28 in Example 37 step 5 with Intermediate 48.
MS (ESI): 465 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.29 (br s, 1 H), 9.92 (s, 1 H), 8.78 (s, 1 H), 7.94 (d, 2H), 7.53 (d, 2H), 7.45 (m, 1 H), 7.37 (m, 2H), 6.60 (br s, 1 H), 6.20 (s, 1 H), 3.40 (m, 2H), 2.88 (m, 2H), 2.32 (m, 2H), 2.13 (s, 3H), 1.31 (s, 9H).
(2) 4-tert-Butyl-N-(3-r6-(1-methanesulfonyl-1.2.3.6-tetrahvdro-pyridin-4-vn-7H- pyrrolor2,3-dlpyrimidin-4-yll-2-methyl-phenyl)-benzamide
Figure imgf000088_0002
To a solution of Intermediate 39 (80 mg, 0.172 mmol) and TEA (0.048 ml, 0.344 mmol) in DCM was added methanesulfonyl chloride (0.021 ml, 0.258 mmol) dropwise. The resulting mixture was stirred at r.t. for 16 hrs, then quenched with water, and extracted with EtOAc. The organic layer was dried with Na2S04 and filtered. The solvents were removed in vacuo, and the crude product was purified by flash chromatography (silica gel, EtOAc/MeOH/NHs gradient) to yield Example 44. MS (ESI): 544 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.34 (br s, 1 H), 9.92 (s, 1 H), 8.81 (s, 1 H), 7.94 (d, 2H), 7.54 (d, 2H), 7.46 (m, 1 H), 7.37 (m, 2H), 6.60 (br s, 1 H), 6.31 (s, 1 H), 3.92 (m, 2H), 3.36 (m, 2H), 2.93 (s, 3H), 2.61 (m, 2H), 2.14 (s, 3H), 1.32 (s, 9H).
Example 45
4-(4-r3-(6-tert-Butyl-1-oxo-3,4-dihvdro-1 H-isoquinolin-2-yl)-2-hvdroxymethyl-phenvn Pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000089_0001
(1 ) Acetic acid 2-(6-tert-butyl-1-oxo-3.4-dihvdro-1 H-isoguinolin-2-vn-6-(4.4.5.5- tetramethyl-H ,3,21dioxaborolan-2-yl)-benzyl ester, Intermediate 40
Figure imgf000089_0002
Intermediate 40 was prepared analogue to Intermediate 4 by replacing 1-bromo-5- fluoro-2-methyl-3-nitro-benzene with acetic acid 2-bromo-6-(6-tert-butyl-1-oxo-3,4- dihydro-1 H-isoquinolin-2-yl)-benzyl ester (WO2010/000633). (2) 4-(4-r3-(6-tert-Butyl-1-oxo-3.4-dihvdro-1 H-isoauinolin-2-vn-2-hvdroxymethyl- phenyll-7H-pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carbo acid dimethylamide
Figure imgf000090_0001
Example 45 was prepared analogue to Intermediate 6 by replacing Intermediate 5 with Intermediate 40.
MS (ESI): 579 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 8.83 (s, 1 H), 7.88 (m, 1 H), 7.68 (m, 1 H), 7.58 (m, 1 H), 7.44 (m, 1 H), 7.42 (m, 1 H), 6.61 (s, 1 H), 6.58 (s, 1 H), 5.24 (m, 1 H), 4.39 (m, 2H), 4.02 (m, 1 H), 3.92 (m, 2H), 3.89 (m, 1 H), 3.36 (m, 2H), 3.29 (m, 1 H), 3.13 (m, 1 H), 2.78 (s, 6H), 2.55 (m, 2H), 1.33 (s, 9H).
Example 46
4-f4-r3-(6-Cvclopropyl-1-oxo-3,4-dihvdro-1 H-isoquinolin-2-vn-2-hvdroxymethyl-phenyll-
7H-pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000090_0002
(1 ) Acetic acid 2-(6-cvclopropyl-1-oxo-3,4-dihvdro-1 H-isoquinolin-2-yl)-6-(4,4,5,5- tetramethyl-ri,3,21dioxaborolan-2-yl)-benzyl ester, Intermediate 41
Figure imgf000091_0001
Intermediate 41 was prepared analogue to Intermediate 4 by replacing 1-bromo-5- fluoro-2-methyl-3-nitro-benzene with acetic acid 2-bromo-6-(6-cyclopropyl-1-oxo-3,4- dihydro-1 H-isoquinolin-2-yl)-benzyl ester (WO2010/000633).
(2) 4-f4-r3-(6-Cvclopropyl-1-oxo-3,4-dihvdro-1 H-isoquinolin-2-vn-2-hvdroxymethyl- phenyll-7H-pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000091_0002
Example 46 was prepared analogue to Intermediate 6 by replacing Intermediate 5 with Intermediate 41.
MS (ESI): 563 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 8.81 (s, 1 H), 7.80 (d, 1 H), 7.66 (d, 1 H), 7.56 (t, 1 H), 7.50 (d, 1 H), 7.09 (m, 1 H), 7.08 (m, 1 H), 6.60 (s, 1 H), 6.54 (s, 1 H), 5.15 (m, 1 H), 4.36 (m, 2H), 3.98 (m, 1 H), 3.91 (m, 2H), 3.89 (m, 1 H), 3.35 (m, 2H), 3.24 (m, 1 H), 3.09 (m, 1 H), 2.77 (s, 6H), 2.55 (m, 2H), 2.00 (m, 1 H), 1.04 (m, 2H), 0.78 (m, 2H). Example 47
4-(4-{2-Hvdroxymethyl-3-r6-(1-hvdroxy-1-methyl-ethyl)-1-oxo-3,4-dihydro-1 H
2-yll-phenyl)-7H-pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihydro-2H-pyridine- acid dimethylamide
Figure imgf000092_0001
(1 ) 2-Bromo-6-r6-(1-hvdroxy-1-methyl-ethyl)-1-oxo-3^-dihydro-1 H-isoquinolin-2-yll- benzaldehyde, Intermediate 42
Figure imgf000092_0002
To a mixture of 2,6-dibromo-benzaldehyde (1.60 g, 6.06 mmol), Pd2(dba)3 (222 mg, 0.243 mmol), xantphos (210 mg, 0.364 mmol), and Cs2C03 (2.77 g, 8.49 mmol) in dioxane (60 ml) under argon was added 6-(1-hydroxy-1-methyl-ethyl)-3,4-dihydro-2H- isoquinolin-1-one (US2009/0306041 ) (995 mg, 4.85 mmol). The resulting mixture was refluxed overnight. After cooling down to r.t., water was added, and the mixture was extracted three times with EtOAc. The combined organic layers were dried, filtered, and evaporated to dryness. The crude product was purified by flash chromatography (silica gel, cyclohexane/EtOAC gradient) to yield Intermediate 42. (2) 2-(3-Bromo-2-hvdroxymethyl-phenyl)-6-(1-hvdroxy-1-methyl-ethyl)-3,4-dihydro- 2H-isoquinolin-1-one, Intermediate 43
Figure imgf000093_0001
To a solution of Intermediate 42 (320 mg, 0.824 mmol) in THF (10 ml) under argon atmosphere was added lithium triethylborohydride (1 M in THF, 0.989 ml, 0.989 mmol) dropwise. The resulting mixture was stirred for 2 hrs at r.t. before adding sat. aqueous NaHC03 solution (5 ml). Additional water was added and the mixture was extracted three times with EtOAc. The combined organic layers were dried, filtered, and evaporated to dryness. The crude product was purified by flash chromatography (silica gel, cyclohexane/EtOAC gradient) to yield Intermediate 43.
MS (ESI): 390 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 7.83 (d, 1 H), 7.63 (d, 1 H), 7.48 (m, 1 H), 7.47 (s, 1 H), 7.36 (m, 1 H), 7.34 (m, 1 H), 5.16 (s, 1 H), 4.92 (m, 1 H), 4.53 (m, 1 H), 4.46 (m, 1 H), 3.96 (m, 1 H), 3.77 (m, 1 H), 3.25 (m, 1 H), 3.07 (m, 1 H), 1.45 (s, 6H).
(3) Acetic acid 2-bromo-6-r6-(1-hvdroxy-1-methyl-ethyl)-1-oxo-3,4-dihydro-1 H- isoquinolin-2-yll-benzyl ester, Intermediate 44
Figure imgf000093_0002
To a solution of Intermediate 43 (238 mg, 0.610 mmol) in DCM (6 ml) were added TEA (0.085 ml, 0.610 mmol), DMAP (7.5 mg, 0.061 mmol), and acetic anhydride (62 mg, 0.610 mmol). The resulting mixture was stirred for 24 hrs at r.t. The solvents were removed in vacuo and the crude product was purified by flash chromatography (silica gel, cyclohexane/EtOAC gradient) to yield Intermediate 44.
MS (ESI): 432 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 7.82 (d, 1 H), 7.70 (m, 1 H), 7.47 (m, 3H), 7.44 (m, 1 H), 5.16 (s, 1 H), 5.1 1 (m, 1 H), 5.04 (m, 1 H), 4.03 (m, 1 H), 3.67 (m, 1 H), 3.18 (m, 1 H), 3.09 (m, 1 H), 1.97 (s, 3H), 1.45 (s, 6H). (4) Acetic acid 2-r6-(1-hvdroxy-1-methyl-ethvn-1-oxo-3,4-dihydro-1 H-isoquinolin-2- yll-6-(4,4,5,5-tetramethyl-ri ,3,21dioxaborolan-2-yl)-benzyl ester, Intermediate 45
Figure imgf000094_0001
Intermediate 45 was prepared analogue to Intermediate 4 by replacing 1-bromo-5- fluoro-2-methyl-3-nitro-benzene with Intermediate 44.
MS (ESI): no peaks , 1H-NMR (DMSO-d6): δ (ppm) 7.82 (d, 1 H), 7.66 (m, 1 H), 7.48-7.46 (m, 4H), 5.29 (d, 1 H), 5.15 (s, 1 H), 4.99 (d, 1 H), 4.02 (m, 1 H), 3.65 (m, 1 H), 3.16 (m, 1 H), 3.09 (m, 1 H), 1.92 (s, 3H), 1.45 (s, 6H), 1.31 (s, 12H).
(5) 4-(4-(2-Hvdroxymethyl-3-r6-(1-hvdroxy-1-methyl-ethvn-1-oxo-3,4-dihvdro-1 H- isoquinolin-2-yll-phenyl)-7H-pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1- carboxylic acid dimethylamide
Figure imgf000094_0002
Example 47 was prepared analogue to Intermediate 6 by replacing Intermediate 5 with Intermediate 45, followed by basic removal of the acetate protecting group with LiOH in MeOH/water.
MS (ESI): 581 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.47 (br s, 1 H), 8.82 (s, 1 H), 7.86 (d, 1 H), 7.66 (d, 1 H), 7.57 (t, 1 H), 7.49 (m, 3H), 6.60 (s, 1 H), 6.55 (s, 1 H), 5.21 (m, 1 H), 5.16 (s, 1 H), 4.37 (m, 2H), 4.02 (m, 1 H), 3.90 (m, 2H), 3.35-3.25 (m, 3H), 3.13 (m, 1 H), 2.77 (s, 6H), 2.55 (m, 2H), 1.46 (s, 6H). Example 48
4-tert-Butyl-N-(3-r6-(3,6-dihvdro-2H-pyran-4-yl)-7H-pyrrolor2,3-dlpyrimidi
hydroxymethyl-phenvD-benzamide
Figure imgf000095_0001
Example 48 was prepared analogue to Intermediate 29 by replacing Intermediate 1 with Intermediate 11 , followed by removal of the TBDPS protecting group with TBAF in THF.
MS (ESI): 483 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.44 (br s, 1 H), 10.52 (s, 1 H), 8.82 (s, 1 H), 8.21 (d, 1 H), 7.90 (d, 2H), 7.58 (d, 2H), 7.50 (t, 1 H), 7.35 (d, 1 H), 6.64 (br s, 1 H), 6.42 (s, 1 H), 5.91 (m, 1 H), 4.68 (d, 2H), 4.28 (m, 2H), 3.81 (m, 2H), 2.46 (m, 2H), 1.32 (s, 9H).
Example 49
4-tert-Butyl-N-(3-r6-(3.6-dihvdro-2H-pyran-4-vn-7H-pyrrolor2.3-dlpyrimidin-4-yll-5-fluoro-
2-methyl-phenyl)-benzamide
Figure imgf000095_0002
Example 49 was prepared analogue to Intermediate 6 by replacing Intermediate 3 with Intermediate 11 and Intermediate 5 with Intermediate 38.
MS (ESI): 485 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.44 (br s, 1 H), 9.97 (s, 1 H), 8.82 (s, 1 H), 7.94 (d, 2H), 7.57 (d, 2H), 7.50(m, 1 H), 7.20 (m, 1 H), 6.64 (s, 1 H), 6.32 (s, 1 H), 4.28 (s, 2H), 3.80 (m, 2H), 2.47 (m, 2H), 2.1 1 (s, 3H), 1.32 (s, 9H).
Example 50
N-(3-r6-(3.6-Dihvdro-2H-pyran-4-vn-7H-pyrrolor2.3-dlpyrimidin-4-yl1-5-fluoro-2-methyl- phenyl)-4-dimethylamino-benzamide
Figure imgf000096_0001
(1 ) N-(3-Bromo-5-fluoro-2-methyl-phenyl)-4-dimethylamino-benzamide,
Intermediate 46
Figure imgf000096_0002
Intermediate 46 was prepared analogue to Intermediate 34 by replacing Intermediate
33 with 4-dimethylamino-benzoyl chloride.
MS (ESI): 353 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 9.77 (br s, 1 H), 7.85 (d, 2H), 7.45 (d, 1 H), 7.31 (d, 1 H), 6.77 (d, 2H), 3.00 (s, 6H), 2.24 (s, 3H). (2) 4-Dimethylamino-N-r5-fluoro-2-methyl-3-(4^,5,5-tetramethyl-ri ,3,21dioxaborolan- 2-yl)-phenyll-benzamide, Intermediate 47
Figure imgf000097_0001
Intermediate 47 was prepared analogue to Intermediate 4 by replacing 1-bromo-5- fluoro-2-methyl-3-nitro-benzene with Intermediate 46.
(3) N-(3-r6-(3.6-Dihvdro-2H-pyran-4-vn-7H-pyrrolor2.3-dlpyrimidin-4-yl1-5-fluoro-2- methyl-phenyl)-4-dimethylamino-benzamide
Figure imgf000097_0002
Example 50 was prepared analogue to Example 49 by replacing Intermediate 38 with Intermediate 47.
MS (ESI): 472 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.43 (br s, 1 H), 9.64 (s, 1 H), 8.82 (s, 1 H), 7.88 (d, 2H), 7.46 (d, 1 H), 7.15 (d, 1 H), 6.78 (d, 2H), 6.64 (s, 1 H), 6.31 (s, 1 H), 4.28 (s, 2H), 3.80 (m, 2H), 3.00 (s, 6H), 2.47 (m, 2H), 2.10 (s, 3H). Example 51
4-tert-Butyl-N-(3-r6-(3,6-dihvdro-2H-thiopyran-4-yl)-7H-pyrrolor2,3-dlpyrim
methyl-phenvD-benzamide
Figure imgf000098_0001
( 1 ) 4-tert-Butyl-N-r2-methyl-3-(4.4.5.5-tetramethyl-n .3.21dioxaborolan-2vn-phenyll- benzamide, Intermediate 48
Figure imgf000098_0002
To a solution of 2-methyl-3-(4,4,5,5-tetramethyl-[1 ,3,2]dioxaborolan-2-yl)-phenylamine (370.0 g, 1.58 mol) in DCM (4 I) and triethylamin (330 ml, 2.38 mol), 4-tert-butylbenzoyl chloride (290 ml, 1.58 mol) was added dropwise over 25 minutes at 0°C. The mixture was stirred at room temperature for 1 hour. The mixture was washed with water, saturated sodium hydrogen carbonate solution and brine. The organic layer was dried over sodium sulfate and evaporated. The residue was taken up in heptane (1 I) and stirred at room temperature for 1 hour. The solid was filtered off and dried to afford Intermediate 48 as a beige solid.
MS (ESI): 394 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 9.80 (s, 1 H), 7.92 (d, 2H), 7.56 (d, 2H), 7.38 (m, 2H), 7.20 (m, 1 H), 2.36 (s, 3H), 1.31 (s, 21 H). (2) 4-tert-Butyl-N-(3-r6-(3.6-dihvdro-2-H-thioDyran-4-vn-7H-Dyrrolor2.3-dlDyrimidin yll-2-methyl-phenyl)-benzamide
Figure imgf000099_0001
For the Suzuki coupling between chloride Intermediate 15 (0.25 g, 0.99 mmol) and the boronic ester Intermediate 48 (0.78 g, 1.98 mmol) the same protocol was used as described in Example 1 step 1 to afford Example 51 as a beige solide.
MS (ESI): 483 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.33 (br s, 1 H), 9.91 (s, 1 H), 8.80 (s, 1 H), 7.93 (d, 2H), 7.59 (m, 1 H), 7.55 (d, 2H), 7.38 (m, 2H), 6.76 (m, 1 H), 6.29 (s, 1 H), 3.35 (m, 2H), 2.82 (m, 2H), 2.64 (m, 2H), 2.14 (s, 3H), 1.32 (s, 9H).
Example 52
4-tert-Butyl -N-(2-methyl-3-r6-(3.6-dihvdro-1-oxido-2H-thiopyran-4-vn-7H-pyrrolor2.3- dlpyrimidin-4-vH-phenyl)-benzamide.
Figure imgf000099_0002
Compound of Example 51 (240 mg, 0.50 mmol) was dissolved in acetic acid (5 ml) and hydrogen peroxide (0.051 ml, 0.50 mmol) was added. The mixture was stirred at room temperature for 4 hours. The mixture was treated with sodium hydrogen sulfite solution 10 % (10 ml) for 10 minutes, diluted with water, basified with 2N sodium hydroxide solution and extracted with EtOAc. The organic layer was washed with brine (2x), dried over sodium sulfate, filtered and evaporated. The residue was purified by flash chromatography on silica (EtOAc to EtOAc/MeOH/NH4OH 9:1 :0.1 to EtOAc/MeOH/NH4OH4 8:2:0.2) to afford the compound Example 52 as a beige solid. MS (ESI): 499 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.45 (br s, 1 H), 9.95 (s, 1 H), 8.84 (s, 1 H), 7.96 (d, 2H), 7.57 (d, 2H), 7.52 (m, 1 H), 7.41 (m, 2H), 6.49 (br s, 1 H), 6.42 (s, 1 H), 3.72 (m, 1 H), 3.49 (m, 1 H), 3.15 (m, 1 H), 2.96 (m, 1 H), 2.89 (m, 1 H), 2.77 (m, 1 H), 2.17 (s, 3H), 1.34 (s, 9H).
Example 53
4-tert-Butyl -N-(3-r6-(3.6-dihvdro-1.1-dioxido-2H-thiopyran-4-vn-7H-pyrrolor2.3- dlpyrimidin-4-yll-2-methyl-phenyl)-benzamide.
Figure imgf000100_0001
Compound of Example 51 (250 mg, 0.51 mmol) was dissolved in DCM (5 ml), then trifluoro acetic acid (5 ml) and hydrogen peroxide (0.079 ml, 0.78 mmol) were added. The mixture was stirred at room temperature for 2 hours. The mixture was treated with sodium hydrogen sulfite solution 10 % (10 ml) for 10 minutes, diluted with water, basified with 2N sodium hydroxide solution and extracted with EtOAc. The organic layer was washed with brine (2x), dried over sodium sulfate, filtered and evaporated. The residue was purified by flash chromatography on silica (EtOAc to EtOAc/MeOH/NH4OH 98:2:0.2) to afford the compound Example 53 as a beige solid.
MS (ESI): 515 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.47 (br s, 1 H), 9.93 (s, 1 H), 8.84 (s, 1 H), 7.94 (d, 2H), 7.60 (m, 1 H), 7.56 (d, 2H), 7.49 (m, 1 H), 7.40 (m, 1 H), 6.48 (m, 1 H), 6.43 (s, 1 H), 4.00 (m, 2H), 3.36 (m, 2H), 3.07 (m, 2H), 2.16 (s, 3H), 1.33 (s, 9H). Example 54
4 ert-Butyl-N-(3-r6-(1 ^-dioxa-spiror4.5ldec-7-en-8-vn-7H-pyrrolor2,3-dlpyrim
hydroxymethyl-phenvD-benzamide
Figure imgf000101_0001
Example 54 was prepared analogue to Intermediate 29 by replacing Intermediate 1 with Intermediate 17, followed by removal of the TBDPS protecting group with TBAF in THF.
MS (ESI): 539 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.34 (br s, 1 H), 10.51 (s, 1 H), 8.80 (s, 1 H), 8.21 (d, 1 H), 7.90 (d, 2H), 7.59 (d, 2H), 7.49 (t, 1 H), 7.33 (d, 1 H), 6.48 (m, 1 H), 6.39 (s, 1 H), 5.90 (m, 1 H), 4.67 (m, 2 H), 3.91 (m, 4H), 2.57 (m, 2H), 2.42 (m, 2H), 1.79 (m, 2H), 1.32 (s, 9H).
Example 55
4-tert-Butyl-N-f3-r6-(4-hvdroxy-cvclohex-1-enyl)-7H-pyrrolor2,3-dlpyrimidin-4-yll-2- hydroxymethyl-phenvD-benzamide
Figure imgf000101_0002
(1 ) 4 ert-Butyl-N-(2-hvdroxymethyl-3-r6-(4-oxo-cvclohex-1-enyl)-7H-pyrrolor2,3- dlpyrimidin-4-yll-phenyl)-benzamide, Intermediate 49
Figure imgf000102_0001
Intermediate 49 was prepared analogue to Intermediate 19 by replacing Example 27 with Example 54.
MS (ESI): 495 [M+H]+
(2) 4-tert-Butyl-N-(3-r6-(4-hvdroxy-cvclohex-1-enyl)-7H-pyrrolor2,3-dlpyrimidin-4-yll-
2-hydroxymethyl-phenyl)-benzamide
Figure imgf000102_0002
To a solution of Intermediate 49 (80 mg, 0.162 mmol) in MeOH/DCM (1 : 1 ) were added cerium(lll) chloride heptahydrate (66.3 mg, 0.178 mmol) and NaBH4 (6.1 mg, 0.162 mmol). The resulting mixture was stirred at r.t. for 2 hrs. Water was added, and the mixture was extracted three times with DCM/isopropanol (3:1 ). The combined organic layers were dried, filtered, and evaporated to dryness. The resulting residue was purified using flash chromatography (silica gel, EtOAc/MeOH/NH3 gradient) to yield Example 55. MS (ESI): 497 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.29 (br s, 1 H), 10.51 (s, 1 H), 8.79 (s, 1 H), 8.19 (d, 1 H), 7.90 (d, 2H), 7.59 (d, 2H), 7.49 (t, 1 H), 7.34 (d, 1 H), 6.49 (m, 1 H), 6.35 (s, 1 H), 5.89 (m, 1 H), 4.72 (m, 1 H), 4.67 (m, 2 H), 3.80 (m, 1 H), 2.65-2.35 (m, 3H), 2.1 1 (m, 1 H), 1.84 (m, 1 H), 1.59 (m, 1 H), 1.33 (s, 9H).
Example 56
4-tert-Butyl-N-(3-r6-(4-dimethylamino^
hvdroxymethyl-phenvD-benzamide
Figure imgf000103_0001
To a solution of Intermediate 49 (100 mg, 0.202 mmol) in DCM/MeOH/AcOH (4 ml, 100:93:7) was added dimethylamine (91 mg, 2.02 mmol). The mixture was stirred for 15 min at r.t. before adding sodium cyanoborohydride (12.7 mg, 0.202 mmol). The mixture was stirred at r.t. overnight and then quenched with 2N HCI (3 ml). After additional stirring for 30 min water and 2N NaOH were added. The mixture was extracted with EtOAc, and the organic layer was dried, filtered, and evaporated to dryness. The resulting residue was purified using flash chromatography (silica gel, EtOAc/MeOH/NH4OH gradient) to yield Example 56.
MS (ESI): 524 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.31 (br s, 1 H), 10.51 (s, 1 H), 8.79 (s, 1 H), 8.19 (m, 1 H), 7.90 (d, 2H), 7.59 (d, 2H), 7.49 (t, 1 H), 7.34 (d, 1 H), 6.57 (m, 1 H), 6.35 (s, 1 H), 5.89 (m, 1 H), 4.66 (m, 2H), 2.60-2.15 (m, 5H), 2.27 (m, 3H), 1.99 (m, 1 H), 1.48 (m, 1 H), 1.31 (s, 9H). Example 57
4-(4-{3-r(3-tert-Butoxy-azetidine-1-carbonyl)-aminol-5-fl^
Pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000104_0001
A mixture of Intermediate 6 (50 mg, 0.127 mmol), 4-nitrophenylchloroformate (25.6 mg, 0.127 mmol), pyridine (12 mg, 0.152 mmol), and DMAP (1.5 mg, 0.013 mmol) in THF (2 ml) was stirred ar r.t. for 5 hrs. Then additional 4-nitrophenylchloroformate (25.6 mg, 0.127 mmol) was added and stirring at r.t. was continued for 72 hrs. Then pyridine (100 mg, 1.27 mmol) and 3-tert-butoxy-azetidine hydrochloride (US2009/0105209) (105 mg, 0.634 mmol) were added and the mexture was stirred for 1 hr. The mixture was diluted into EtOAc and aqueous NH4OH solution (2 M). The organic layer was washed with brine, dried with Na2S04, filtered, and evaporated to dryness. The crude product was purified by flash chromatography (siliga gel, EtOAc/MeOH gradient) to afford Example 57 as beige solid.
MS (ESI): 550 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.45 (br s, 1 H), 8.80 (s, 1 H), 7.97 (br s, 1 H), 7.44 (m, 1 H), 7.00 (m, 1 H), 6.57 (br s, 1 H), 6.27 (s, 1 H), 4.52 (m, 1 H), 4.17 (m, 2H), 3.87 (m, 2H), 3.75 (m, 2H), 3.30 (m, 2H), 2.74 (s, 6H), 2.49 (m, 2H), 2.03 (s, 3H), 1.13 (s, 9H).
Example 58
4-(4-{5-Fluoro-3-r(3-isopropoxy-azetidine-1-carbonyl)-aminol-2-methyl-phenyl)-7H Pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000105_0001
Example 58 was prepared analogue to Example 57 by replacing 3-tert-butoxy-azetidine hydrochloride with 3-isopropoxy-azetidine hydrochloride.
MS (ESI): 536 [M+H]+ , 1H-NMR (CDCI3): δ (ppm) 12.24 (br s, 1 H), 8.86 (s, 1 H), 7.78 (m, 1 H), 6.93 (m, 1 H), 6.49 (m, 1 H), 6.33 (s, 1 H), 6.28 (m, 1 H), 4.39 (m, 1 H), 4.24 (m, 2H), 4.03 (m, 2H), 3.97 (m, 2H), 3.62 (m, 1 H), 3.47 (m, 2H), 2.86 (s, 6H), 2.62 (m, 2H), 2.1 1 (s, 3H), 1.16 (d, 6H).
Example 59
5-Fluoro-1 ,3-dihydro-isoindole-2-carboxylic acid (3-r6-(1-dimethylcarbamoyl-1 ,2,3,6- tetrahvdro-pyridin-4-yl)-7H-pyrrolor2,3-dlpyrimidin-4-yll-5-fluoro-2-methyl-phenyl)-am
Figure imgf000105_0002
Example 59 was prepared analogue to Example 57 by replacing 3-tert-butoxy-azetidine hydrochloride with 5-fluoro-2,3-dihydro-1 H-isoindole.
MS (ESI): 558 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.40 (br s, 1 H), 8.81 (s, 1 H), 7.96 (s, 1 H), 7.56 (m, 1 H), 7.41 (m, 1 H), 7.26 (m, 1 H), 7.14 (m, 1 H), 7.04 (m, 1 H), 6.58 (s, 1 H), 5.30 (s, 1 H), 4.78 (m, 4H), 3.89 (m, 2H), 3.34 (m, 2H), 2.76 (s, 6H), 2.52 (m, 2H), 2.1 1 (s, 3H).
Example 60
4-r4-(5-Fluoro-2-methyl-3-(r3-(2,2,2 rifluoro-1 rifluoromethyl-ethoxy)-azetidin
carbonyll-amino)-phenyl)-7H-pyrrolor2,3-dlpyrimidin-6-yll-3,6-dihydro-2H-py
carboxylic acid dimethylamide
Figure imgf000106_0001
Example 60 was prepared analogue to Example 57 by replacing 3-tert-butoxy-azetidine hydrochloride with 3-(2,2,2-trifluoro-1-trifluoromethyl-ethoxy)-azetidine hydrochloride (WO2009/077334).
MS (ESI): 644 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.35 (br s, 1 H), 8.76 (s, 1 H), 8.10 (s, 1 H), 7.39 (m, 1 H), 7.00 (m, 1 H), 6.54 (m, 1 H), 6.23 (s, 1 H), 5.63 (m, 1 H), 4.75 (m, 1 H), 4.25 (m, 2H), 3.87 (m, 2H), 3.85 (m, 2H), 3.82 (m, 2H), 2.72 (s, 6H), 2.46 (m, 2H), 2.01 (s, 3H).
Example 61
3-tert-Butoxy-azetidine-1-carboxylic acid {3-r6-(3,6-dihvdro-2H-pyran-4-yl)-7H-
Pyrrolor2,3-dlpyrimidin-4-yll-5-fluoro-2-methyl-phenyl)-amide
Figure imgf000107_0001
Example 61 was prepared analogue to Example 57 by replacing Intermediate 6 with Intermediate 12.
MS (ESI): 480 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.40 (br s, 1 H), 8.80 (s, 1 H), 7.95 (s, 1 H), 7.43 (m, 1 H), 7.00 (m, 1 H), 6.62 (br s, 1 H), 6.26 (s, 1 H), 4.52 (m, 1 H), 4.27 (m, 2H), 4.18 (m, 2H), 3.79 (m, 2H), 3.75 (m, 2H), 2.44 (m, 2H), 2.04 (s, 3H), 1.13 (s, 9H).
Example 62
4-(4-(3-r(3 ert-Butoxy-azetidine-1-carbonyl)-aminol-4-fluoro-phenyl)-7H-pyrrolor2,3- dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000107_0002
Example 62 was prepared analogue to Example 57 by replacing Intermediate 6 with Intermediate 10. MS (ESI): 536 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.36 (br s, 1 H), 8.77 (s, 1 H), 8.49 (m, 1 H), 8.33 (s, 1 H), 7.90 (m, 1 H), 7.39 (m, 1 H), 6.88 (s, 1 H), 6.59 (s, 1 H), 4.53 (m, 1 H), 4.21 (m, 2H), 3.91 (m, 2H), 3.78 (m, 2H), 3.37 (m, 2H), 2.77 (s, 6H), 2.60 (m, 2H), 1.14 (s, 9H).
Example 63
4-(4-(3-r(3 ert-Butoxy-azetidine-1-carbonyl)-aminol-4-fluoro-2-methyl-phenyl)-7H- Pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000108_0001
(1 ) 4-r4-(3-Amino-4-fluoro-2-methyl-phenvn-7H-pyrrolor2.3-dlpyrimidin-6-yll-3.6- dihydro-2H-pyridine-1-carboxylic acid dimethylamide, Intermediate 50
Figure imgf000108_0002
Intermediate 10 was prepared analogue to Intermediate 6 by replacing Intermediate 5 with 5-fluoro-2-methyl-3-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)aniline.
MS (ESI): 395 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.28 (br s, 1 H), 8.75 (s, 1 H), 7.03 (m, 1 H), 6.68 (m, 1 H), 6.54 (s, 1 H), 6.25 (s, 1 H), 5.00 (br s, 2H), 3.88 (s, 2H), 3.32 (m, 2H), 2.75 (s, 6H), 2.50 (m, 2H), 2.03 (s, 3H). (2) 4-(4-(3-r(3 ert-Butoxy-azetidine-1-carbonvn-aminol-4-fluoro-2-methyl-phenylV 7H-pyrrolor2,3-dlpyrimidin-6-yl)-3,6-dihvdro-2H-pyridine-1-carboxylic acid dimethylamide
Figure imgf000109_0001
Example 62 was prepared analogue to Example 57 by replacing Intermediate 6 with Intermediate 50.
MS (ESI): 550 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.36 (br s, 1 H), 8.79 (s, 1 H), 8.04 (m, 1 H), 7.37 (m, 1 H), 7.21 (m, 1 H), 6.57 (m, 1 H), 6.25 (s, 1 H), 4.54 (m, 1 H), 4.14 (m, 2H), 3.89 (m, 2H), 3.70 (m, 2H), 3.32 (m, 2H), 2.75 (s, 6H), 2.50 (m, 2H), 2.13 (s, 3H), 1.14 (s, 9H).
Example 64
4-tert-Butyl-N-(3-r6-(3.6-dihvdro-2H-pyran-4-vn-7H-pyrrolor2.3-dlpyrimidin-4-yl1-2- methyl-phenvD-benzamide
Figure imgf000109_0002
(1 ) 4 ert-Butyl-N-r2-methyl-3-(7H-pyrrolor2,3-dlpyrimidin-4-yl)-phenyll-be
Intermediate 51
Figure imgf000110_0001
Intermediate 51 was prepared analogue to Intermediate 6 by replacing Intermediate 3 with 6-chloro-7-deazapurine and Intermediate 5 with Intermediate 48.
MS (ESI): 385 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.22 (br s, 1 H), 9.94 (s, 1 H), 8.82 (s, 1 H), 7.94 (d, 2H), 7.60 (m, 1 H), 7.58 (d, 2H), 7.37 (m, 1 H), 6.30 (m, 1 H), 2.12 (s, 3H), 1.31 (s, 9H).
(2) N-r3-(7-Benzenesulfonyl-7H-pyrrolor2,3-dlpyrimidin-4-yl)-2-methyl-phenyll-4-tert- butyl-benzamide, Intermediate 52
Figure imgf000110_0002
To a solution of Intermediate 51 (7.51 g, 19.54 mmol) in THF (500 ml) at 0°C was added NaH (1.172 g, 29.3 mmol) portionwise. The mixture was stirred at 0°C for 3 hrs, then benzenesulfonyl chloride (3.70 ml, 25.4 mmol) was added and stirring was continued at r.t. overnight. The reaction mixture was quenched with sat. aqueous NH4CI solution, and extracted with EtOAc. The organic layer was dried, filtered, and evaporated to dryness. Purification by flash chromatography (silica gel, cyclohexane/EtOAc gradient) yielded Intermediate 52.
MS (ESI): 525 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 9.97 (s, 1 H), 9.10 (s, 1 H), 8.21 (d, 2H), 8.05 (d, 1 H), 7.92 (d, 2H), 7.78 (t, 1 H), 7.69 (t, 2H), 7.60 (m, 1 H), 7.53 (d, 2H), 7.51 (d, 1 H), 7.37 (t, 1 H), 7.32 (d, 1 H), 6.62 (d, 1 H), 2.12 (s, 3H), 1.31 (s, 9H).
(3) N-r3-(7-Benzenesulfonyl-6-bromo-7H-pyrrolor2,3-dlpyrimidin-4-yl)-2-methyl- phenyll-4-tert-butyl-benzamide, Intermediate 53
Figure imgf000111_0001
To a solution of Intermediate 52 (4.26 g, 8.12 mmol) in THF (150 ml) at -78°C was added LDA (1.5 M in THF, 16.24 ml, 24.36 mmol) slowly. The resulting mixture was stirred for 1.5 hrs at -78°C before adding a solution of 1 ,2-dibromo-tetrachloroethane (3.97 g, 12.18 mmol) in THF (1 ml). Stirring was continued at -78°C for 2 hrs, then the reaction mixture was quenched by addition of sat. aqueous NH4CI solution and warmed up to r.t. The mixture was extracted with EtOAc and the organic layer was dried, filtered, and evaporated to dryness. Purification by flash chromatography (silica gel, cyclohexane/EtOAc gradient) yielded Intermediate 53.
MS (ESI): 603 [M+H]+ (4) N-r3-(6-Bromo-7H-Dyrrolor2.3-dlDyrimi^
benzamide, Intermediate 54
Figure imgf000112_0001
To a solution of Intermediate 53 (3.82 g, 6.33 mmol) in THF (64 ml) was added a solution of potassium tert-butylate in THF (1 M, 9.49 ml, 9.49 mmol) at 0°C. The mixture was stirred at 0°C for 1 hr, then an additional solution of potassium tert-butylate in THF (1 M, 2.00 ml, 2.00 mmol) was added and stirring was continued for an additional 1 hr. The reaction was quenched by addition of sat. aqueous NaHC03 solution, and the mixture was extracted with EtOAc. The organic layer was dried, filtered, and evaporated to dryness. Purification by flash chromatography (silica gel, cyclohexane/EtOAc gradient) yielded Intermediate 54.
MS (ESI): 463 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 13.10 (br s, 1 H), 9.97 (s, 1 H), 8.82 (s, 1 H), 7.93 (d, 2H), 7.54 (d, 1 H), 7.48 (d, 1 H), 7.40-7.32 (m, 2H), 6.41 (s, 1 H), 2.12 (s, 3H), 1.31 (s, 9H).
(5) 4-tert-Butyl-N-(3-r6-(3.6-dihvdro-2H-pyran-4-vn-7H-pyrrolor2.3-dlpyrimidin-4-yl1-
2-methyl-phenyl)-benzamide
Figure imgf000112_0002
Example 64 was prepared analogue to Intermediate 11 by replacing 4-chloro-6-iodo- 7H-pyrrolo[2,3-d]pyrimidine with Intermediate 54.
MS (ESI): 467 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.33 (br s, 1 H), 9.93 (s, 1 H), 8.82 (s, 1 H), 7.94 (d, 2H), 7.54 (d, 2H), 7.48 (m, 1 H), 7.37 (m, 2H), 6.63 (br s, 1 H), 6.25 (s, 1 H), 4.36 (m, 2H), 3.81 (m, 2H), 2.46 (m, 2H), 2.16 (s, 3H), 1.33 (s, 9H).
Example 65
4-tert-Butyl-N-(3-r6-(1.4-dioxa-SDiror4.5ldec-7-en-8-vn-7H-Dyrrolor2.3-dlDyrimidin-4-yll-2- methyl-phenvD-benzamide
Figure imgf000113_0001
Example 65 was prepared analogue to Intermediate 17 by replacing 4-chloro-6-iodo- 7H-pyrrolo[2,3-d]pyrimidine with Intermediate 54.
MS (ESI): 523 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.28 (br s, 1 H), 9.91 (s, 1 H), 8.78 (s, 1 H), 7.94 (d, 2H), 7.54 (d, 2H), 7.49 (m, 1 H), 7.37 (m, 2H), 6.46 (br s, 1 H), 6.23 (s, 1 H), 3.90 (s, 4H), 2.55 (m, 2H), 2.41 (m, 2H), 2.14 (s, 3H), 1.79 (m, 2H), 1.31 (s, 9H).
Example 66
4-tert-Butyl-N-(3-r6-(4-hvdroxy-cvclohex-1-enyl)-7H-pyrrolor2,3-dlpyrimidin-4-yll-2- methyl-phenvD-benzamide
Figure imgf000114_0001
(1 ) 4-tert-Butyl-N-(2-methyl-3-r6-(4-oxo-cvclohex-1-enyl)-7H-pyrrolor2,3-dlpyrimidin- 4-yll-phenyl)-benzamide, Intermediate 55
Figure imgf000114_0002
Intermediate 55 was prepared analogue to Intermediate 19 by replacing Example 27 with Example 65.
MS (ESI): 479 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.35 (br s, 1 H), 9.92 (s, 1 H), 8.78 (s, 1 H), 7.94 (d, 2H), 7.53 (d, 2H), 7.49 (m, 1 H), 7.36 (m, 2H), 6.64 (br s, 1 H), 6.35 (s, 1 H), 3.15 (m, 2H), 2.83 (m, 2H), 2.52 (m, 2H), 2.13 (s, 3H), 1.31 (s, 9H). (2) 4-tert-Butyl-N-(3-r6-(4-hvdroxy-cvclohex-1-enyl)-7H-pyrrolor2,3-dlpyrimidin-4-yl^ 2-methyl-phenvD-benzamide
Figure imgf000115_0001
Example 66 was prepared analogue to Example 28 step 2 by replacing Intermediate 19 in with Intermediate 55.
MS (ESI): 481 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.21 (br s, 1 H), 9.93 (s, 1 H), 8.77 (s, 1 H), 7.94 (d, 2H), 7.53 (d, 2H), 7.43 (m, 1 H), 7.37 (m, 2H), 6.47 (br s, 1 H), 6.18 (s, 1 H), 4.75 (s, 1 H), 3.76 (m, 1 H), 2.44 (m, 2H), 2.35 (m, 2H), 2.14 (s, 3H), 1.83 (m, 2H), 1.31 (s, 9H).
Example 67
4-tert-Butyl-N-(3-r6-(4-dimethylamino-cvclohex-1-enyl)-7H-pyrrolor2,3-dlpyrimidin-4-yll-2- methyl-phenvD-benzamide
Figure imgf000115_0002
Example 67 was prepared analogue to Example 56 by replacing Intermediate 49 with Intermediate 55. MS (ESI): 508 [M+H]+ , 1H-NMR (DMSO-d6): δ (ppm) 12.23 (br s, 1 H), 9.94 (s, 1 H), 8.78 (s, 1 H), 7.94 (d, 2H), 7.53 (d, 2H), 7.48 (m, 1 H), 7.35 (m, 2H), 6.53 (br s, 1 H), 6.21 (s, 1 H), 2.61 (m, 2H), 2.51 (m, 1 H), 2.42 (m, 1 H), 2.40 br s, 6H), 2.24 (m, 1 H), 2.16 (s, 3H), 2.05 (m, 1 H), 1.56 (m, 1 H), 1.28 (s, 9H).
Biological Part
Inhibition of Btk enzymatic activity
The inhibitory activity of the present compounds against Btk was assessed in a biochemical enzyme assay. Assay plates in 384 well format were prepared with 8-point serial dilutions for the test compounds on a Thermo CatX workstation equipped with a Innovadyne Nanodrop Express. The assay plates were prepared by addition of 50 nl per well of compound solution in 90 % DMSO. The kinase reactions were started by stepwise addition of 4.5 μΙ per well of peptide/ATP-solution (4 μΜ FITC-Ahx- TSELKKVVALYDYMPMNAND-NH2, 164 μΜ ATP) in kinase buffer (50mM HEPES, pH 7.5, 1 mM DTT, 0.02% Tween20, 0.02% BSA, 0.6% DMSO, 10 mM beta- glycerophosphate, and 10 μΜ sodium orthovanadate, 18 mM MgCI2, 1 mM MnCI2) and 4.5 μΙ per well of enzyme solution (6.4nM full-lenght human recombinant BTK) in kinase buffer. Kinase reactions were incubated at 30°C for 60 minutes and subsequently terminated by addition of 16 μΙ per well of stop solution (100 mM HEPES pH 7.5, 5 % DMSO, 0.1 % Caliper coating reagent, 10 mM EDTA, and 0.015 % Brij35). Kinase reactions were analyzed on a Caliper LC3000 workstation by separating phosphorylated and unphosphorylated peptides and kinase activities were calculated from the amounts of newly formed phospho-peptide. Inhibition data were calculated by comparison to control reactions without enzyme (100 % inhibition) and without inhibitors (0 % inhibition). The concentration of inhibitor required for 50 % inhibition (IC50) was calculated from the inhibition in response to inhibitor concentrations.
Figure imgf000116_0001
Example 4 0.002
Example 5 0.003
Example 6 0.002
Example 7 0.003
Example 8 0.003
Example 9 0.005
Example 10 0.015
Example 1 1 0.005
Example 12 0.004
Example 13 0.004
Example 14 0.016
Example 15 0.017
Example 16 0.002
Example 17 0.001
Example 18 0.006
Example 19 0.023
Example 20 0.006
Example 21 0.006
Example 22 0.008
Example 23 0.012
Example 24 0.023
Example 25 0.002
Example 26 0.035
Example 27 0.1 10
Example 28 0.046
Example 29 0.013
Example 30 0.006
Example 31 0.003
Example 32 0.007
Example 33 0.004
Example 34 0.034
Example 35 0.014
Example 36 0.004
Example 37 0.026
Example 38 0.027
Example 39 0.013
Example 40 0.019
Example 41 0.023
Example 42 0.030
Example 43 0.140
Example 44 0.017 Example 45 0.008
Example 46 0.017
Example 47 0.002
Example 48 0.007
Example 49 0.099
Example 50 0.047
Example 51 0.047
Example 52 0.024
Example 53 0.064
Example 54 0.024
Example 55 0.004
Example 56 0.002
Example 57 0.007
Example 58 0.016
Example 59 0.003
Example 60 0.008
Example 61 0.006
Example 62 0.002
Example 63 0.002
Example 64 0.015
Example 65 0.009
Example 66 0.012
Example 67 0.008
Inhibition of cellular Btk activity
Alternatively, the present compounds might also be assessed for their capacity to inhibit Btk-dependent FcG receptor-induced IL-8 secretion in human cells. The human myeloid leukemia THP1 cell line (ATCC TIB202) was grown in RPMI 1640 medium
supplemented with 10 % FCS and 15 nM 1 , 25-dihydroxy Vitamin D3 during 4 days before use to induced myeloid differentiation. A sufficient number of tissue-culture grade 384-well plates was coated with human IgG of unknown specificity by incubating overnight at 4°C with 40 μΙ/well of a 50 μg/ml IgG solution in PBS. On the day of the experiment, plates were washed 5 times with 80 μΙ water on a Molecular Devices Aquamax DW4 plate washer. Solutions of the test compounds in 90 % DMSO were added to each well on a Hamilton Microlab Star liquid handling station to 40 μΙ/well tissue culture medium and the total DMSO concentration was adjusted to 0.1 %.
Differentiated THP1 cells were then added in 40 μΙ/well to reach a final density of 5Ό00 cells/well in 80 μΙ culture medium. After 24 hours, IL-8 secretion was measured in the supernatant by the IL-8 HTRF assay following the protocol of the vendor (CisBio international). Inhibition data were calculated by comparison to control cultures without IgG stimulus enzyme (100 % inhibition) and without inhibitors (0 % inhibition). The concentration of inhibitor required for 50 % inhibition (IC50) was calculated from the inhibition in response to inhibitor concentrations.
Figure imgf000119_0001
Inhibition of Btk activity in blood
Alternatively, the inhibitory activity of the present compounds in blood was assessed in the following in vitro B cell activation assay. Whole blood was collected from the abdominal aorta of anaesthetized adult male Lewis rats and was anticoagulated with 100 U/ml sodium heparin. Blood was then diluted to 50 % with high glucose DMEM
(Amimed) supplemented with 100 U/ml penicillin, 100 mg/ml streptomycin, 2 mM L- glutamin, 50 mg/ml dextran 40 and 5% FCS (Fetadone I, Gibco). Then, 190 μΙ prediluted blood was mixed in 96 well U-bottomed microtiter plates (Nunc) with 10 μΙ of serial dilutions of test compounds in DMSO. Cultures were incubated at 37°C, 5% C02 for 1 hour, then 30 μΙ of rat IL-4 (Beckton-Dickinson, final concentration 5 ng/ml) and goat anti-rat IgM (Serotec, final concentration 15 ug/ml) were added and the cultures were incubated for 24 hours. Activation of B cells was measured by flow cytometry after staining for the B cell subset with PE-Cy5-labeled anti-ratCD45RA (Beckton-Dickinson) and for the activation marker CD86 (PE-labeled anti-rat CD86 (Beckton-Dickinson). All staining procedures were performed at RT for 30 min in the dark in 96-deep well V- bottomed microtiter plates (Corning) with BD Lysing Solution (Beckton-Dickinson). Cytometric data was acquired on a FACScalibur flow cytometer (BD Biosciences) and the subpopulation of lymphocytes were gated according to size and granularity and further analyzed for expression of CD45RA and the activation markers. Data for the inhibition of B cell activation were calculated from the percentage of cells positively stained for activation markers within the CD45RA positive population. Inhibition data were calculated by comparison to control cultures without anti-lgM and IL-4 (100 % inhibition) and without inhibitors (0 % inhibition). The concentration of inhibitor required for 50 % inhibition (IC50) was calculated from the inhibition in response to inhibitor concentrations.
Figure imgf000120_0001
Utilities
Based for example upon the biological test results, compounds of the invention may generally be useful in the treatment of an indication selected from:
Autoimmune disorders, inflammatory diseases, allergic diseases, airway diseases, such as asthma and chronic obstructive pulmonary disease (COPD), transplant rejection; diseases in which antibody production, antigen presentation, cytokine production or lymphoid organogenesis are abnormal or are undesirable; including rheumatoid arthritis, systemic onset juvenile idiopathic arthritis (SOJIA), gout, pemphigus vulgaris, idiopathic thrombocytopenic purpura, systemic lupus erythematosus, multiple sclerosis, myasthenia gravis, Sjogren's syndrome, autoimmune hemolytic anemia, anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitides, cryoglobulinemia, thrombotic thrombocytopenic purpura, chronic autoimmune urticaria, allergy (atopic dermatitis, contact dermatitis, allergic rhinitis), atherosclerosis, type 1 diabetes, type 2 diabetes, inflammatory bowel disease, ulcerative colitis, morbus Crohn, pancreatitis,
glomerolunephritis, Goodpasture's syndrome, Hashimoto's thyroiditis, Grave's disease, antibody-mediated transplant rejection (AMR), graft versus host disease, B cell-mediated hyperacute, acute and chronic transplant rejection; thromboembolic disorders, myocardial infarct, angina pectoris, stroke, ischemic disorders, pulmonary embolism; cancers of haematopoietic origin including but not limited to multiple myeloma; a leukaemia; acute myelogenous leukemia; chronic myelogenous leukemia; lymphocytic leukemia; myeloid leukemia; non-Hodgkin lymphoma; lymphomas; polycythemia vera; essential thrombocythemia; myelofibrosis with myeloid metaplasia; and Waldenstroem disease.
In a further embodiment, the therapy is selected from a disease which may be treated by an antagonist of Bruton's tyrosine kinase.
In another embodiment, the invention provides a method of treating a disease which is treated by the modulation of Btk- comprising administration of a therapeutically acceptable amount of a compound of formula (I) or a salt thereof. In a further embodiment, the disease is selected from the afore-mentioned lists
Combinations
The compound of the present invention may be administered either simultaneously with, or before or after, one or more other therapeutic agent. The compound of the present invention may be administered separately, by the same or different route of
administration, or together in the same pharmaceutical composition as the other agents.
The compounds of formula (I) may be administered as the sole active ingredient or in conjunction with, e.g. as an adjuvant to, other drugs e.g. immunosuppressive or immunomodulating agents or other anti-inflammatory agents, e.g. for the treatment or prevention of alio- or xenograft acute or chronic rejection or inflammatory or autoimmune disorders, or a chemotherapeutic agent, e.g a malignant cell anti-proliferative agent. For example, the compounds of formula (I) may be used in combination with a calcineurin inhibitor, e.g. cyclosporin A or FK 506; a mTOR inhibitor, e.g. rapamycin, 40-O-(2- hydroxyethyl)-rapamycin, CCI779, ABT578, AP23573, AP23464, AP23675, AP23841 , TAFA-93, biolimus-7 or biolimus-9; an ascomycin having immunosuppressive properties, e.g. ABT-281 , ASM981 , etc.; corticosteroids; cyclophosphamide; azathioprene; methotrexate; leflunomide; mizoribine; mycophenolic acid or salt; mycophenolate mofetil; 15-deoxyspergualine or an immunosuppressive homologue, analogue or derivative thereof; a PKC inhibitor, e.g. as disclosed in WO 02/38561 or WO 03/82859, e.g. the compound of Example 56 or 70; a JAK3 kinase inhibitor, e.g. N-benzyl-3,4-dihydroxy- benzylidene-cyanoacetamide a-cyano-(3,4-dihydroxy)-]N-benzylcinnamamide
(Tyrphostin AG 490), prodigiosin 25-C (PNU156804), [4-(4'-hydroxyphenyl)-amino-6,7- dimethoxyquinazoline] (WHI-P131 ), [4-(3'-bromo-4'-hydroxylphenyl)-amino-6,7- dimethoxyquinazoline] (WHI-P154), [4-(3',5'-dibromo-4'-hydroxylphenyl)-amino-6,7- dimethoxyquinazoline] WHI-P97, KRX-21 1 , 3-{(3R,4R)-4-methyl-3-[methyl-(7H- pyrrolo[2,3-d]pyrimidin-4-yl)-amino]-piperidin-1-yl}-3-oxo-propionitrile, in free form or in a pharmaceutically acceptable salt form, e.g. mono-citrate (also called CP-690,550), or a compound as disclosed in WO 04/052359 or WO 05/066156; sphingosine-1-phosphate receptor modulators such as FTY720 (fingolimod), or compounds disclosed in WO 2005/000833; immunosuppressive monoclonal antibodies, e.g., monoclonal antibodies to leukocyte receptors, e.g., MHC, CD2, CD3, CD4, CD7, CD8, CD25, CD28, CD40, CD45, CD52, CD58, CD80, CD86 or their ligands; other immunomodulatory compounds, e.g. a recombinant binding molecule having at least a portion of the extracellular domain of CTLA4 or a mutant thereof, e.g. an at least extracellular portion of CTLA4 or a mutant thereof joined to a non-CTLA4 protein sequence, e.g. CTLA4lg (for ex. designated ATCC 68629) or a mutant thereof, e.g. LEA29Y; adhesion molecule inhibitors, e.g. LFA- 1 antagonists, ICAM-1 or -3 antagonists, VCAM-4 antagonists or VLA-4 antagonists; or a chemotherapeutic agent, e.g. paclitaxel, gemcitabine, cisplatinum, doxorubicin or 5- fluorouracil; or an anti-infectious agent. Further combination partners to a compound of formula (I) may be selected from a PI3K inhibitor (e.g. pan, or alpha, beta, gamma, delta selectives), TNF inhibitors, ILI beta inhibitors, IL17 inhibitors, and inhibitors of IL6 or IL receptor.
The terms "co-administration" or "combined administration" or the like as utilized herein are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are not necessarily administered by the same route of administration or at the same time. The term "pharmaceutical combination" as used herein means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients. The term "fixed combination" means that the active ingredients, e.g. a compound of formula (I) and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage. The term "non-fixed combination" means that the active ingredients, e.g. a compound of formula (I) and a co-agent, are both administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific time limits, wherein such administration provides therapeutically effective levels of the 2 compounds in the body of the patient. The latter also applies to cocktail therapy, e.g. the administration of 3 or more active ingredients.
In one embodiment, the invention provides a product comprising a compound of formula (I) and at least one other therapeutic agent as a combined preparation for simultaneous, separate or sequential use in therapy. In one embodiment, the therapy is the treatment of a disease or condition mediated by Btk kinases. Products provided as a combined preparation include a composition comprising the compound of formula (I) and the other therapeutic agent(s) together in the same pharmaceutical composition, or the compound of formula (I) and the other therapeutic agent(s) in separate form, e.g. in the form of a kit.
In one embodiment, the invention provides a pharmaceutical composition comprising a compound of formula (I) and another therapeutic agent(s). Optionally, the
pharmaceutical composition may comprise a pharmaceutically acceptable excipient, as described above.
In one embodiment, the invention provides a kit comprising two or more separate pharmaceutical compositions, at least one of which contains a compound of formula (I). In one embodiment, the kit comprises means for separately retaining said compositions, such as a container, divided bottle, or divided foil packet. An example of such a kit is a blister pack, as typically used for the packaging of tablets, capsules and the like.
The kit of the invention may be used for administering different dosage forms, for example, oral and parenteral, for administering the separate compositions at different dosage intervals, or for titrating the separate compositions against one another. To assist compliance, the kit of the invention typically comprises directions for
administration.
In the combination therapies of the invention, the compound of the invention and the other therapeutic agent may be manufactured and/or formulated by the same or different manufacturers. Moreover, the compound of the invention and the other therapeutic may be brought together into a combination therapy: (i) prior to release of the combination product to physicians (e.g. in the case of a kit comprising the compound of the invention and the other therapeutic agent); (ii) by the physician themselves (or under the guidance of the physician) shortly before administration; (iii) in the patient themselves, e.g. during sequential administration of the compound of the invention and the other therapeutic agent.
Accordingly, the invention provides the use of a compound of formula (I) for treating a disease or condition mediated by Btk kinases, wherein the medicament is prepared for administration with another therapeutic agent. The invention also provides the use of another therapeutic agent for treating a disease or condition mediated by Btk , wherein the medicament is administered with a compound of formula (I).
The invention also provides a compound of formula (I) for use in a method of treating a disease or condition mediated by Btk, wherein the compound of formula (I) is prepared for administration with another therapeutic agent. The invention also provides another therapeutic agent for use in a method of treating a disease or condition mediated by Btk , wherein the other therapeutic agent is prepared for administration with a compound of formula (I). The invention also provides a compound of formula (I) for use in a method of treating a disease or condition mediated by Btk , wherein the compound of formula (I) is administered with another therapeutic agent. The invention also provides another therapeutic agent for use in a method of treating a disease or condition mediated by Btk, wherein the other therapeutic agent is administered with a compound of formula (I).

Claims

1. A compound of formula (I) or a pharmaceutically acceptable salt thereof;
Figure imgf000125_0001
wherein,
R1 is hydrogen, CrC6 alkyi optionally substituted by hydroxy;
R2 is hydrogen or halogen;
R3 is hydrogen or halogen;
R4 is hydrogen,
R5 is phenyl optionally substituted by halogen; SF5; NR6R7; hydroxy; C C6 alkoxy; d- C6 alkenyl; CrC6 alkyi carbonyl; CrC6 alkyi optionally substituted by hydroxy, halogen, or Ci-C6 alkoxy; or C3-C6 cycloalkyi optionally substituted by halogen, hydroxy, or C C6 alkyi optionally substituted by halogen; or
R5 is a 4 - 14 membered mono- or bicyclic heterocyclyl or heteroaryl ring system comprising 1 , 2 or 3 heteroatoms selected from N, S and O that ring being optionally substituted by halogen; hydroxy; CrC6 alkoxy optionally substituted by hydroxy or halogen; or CrC6 alkyi optionally substituted by hydroxy or halogen;
or R4 and R5 together with the atoms to which they are bound form a piperidone ring, optionally comprising an annulated phenyl ring, any such ring being optionally substituted by CrC6 alkyi, CrC6 alkoxy, or C3-C6 cycloalkyi each of which substitution member may optionally be substituted by halogen or hydroxy;
R6 and R7 are independently selected from hydrogen or CrC6 alkyi;
or R6 and R7 together with the nitrogen atom to which they are bound form a 4 - 8 membered saturated azacycloalkane ring, optionally substituted by halogen, hydroxy or
Ci-C6 alkyi; X is O, S(0)n wherein n is 0, 1 or 2, or
Figure imgf000126_0001
wherein q is 2 or 3, and R10 is absent;
or X is CH or N; and R10 is hydrogen, hydroxy, -NR6R7, -CO-R1 1 , -S(0)p-R12 wherein p is 1 or 2,
R 1 1 is Ci-C6 alkyl optionally substituted by hydroxy, cyano, halogen, carboxy or CrC6 alkoxy carbonyloxy; or NR6R7; and
R12 is Ci-C6 alkyl or NR6R7.
2. A compound according to claim 1 or a pharmaceutically acceptable salt thereof , wherein R1 is hydrogen, methyl or hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 is hydrogen; R5 is phenyl substituted by halogen; CrC6 alkoxy; Ci-C6 alkyl optionally substituted by halogen or hydroxy; or C3-C6 cycloalkyl optionally substituted by halogen, hydroxy, or C C6 alkyl optionally substituted by halogen; X is O,
S(0)n wherein n is 0, 1 or 2, or
Figure imgf000126_0002
wherein q is 2 or 3, and R10 is absent; and the remaining variables are as defined in claim 1.
3. A compound according to claim 1 or 2, or a pharmaceutically acceptable salt thereof, wherein R1 is hydrogen, methyl or hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one optionally substituted by C3-C6 cycloalkyl optionally substituted by hydroxy; X is O,
S(0)n wherein n is 0, 1 or 2, or
Figure imgf000126_0003
wherein q is 2 or 3, and R10 is absent; and the remaining variables are as defined in claim 1.
4. A compound according to claim 1 , or a pharmaceutically acceptable salt thereof, wherein R1 is hydrogen, methyl or hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 is hydrogen; R5 is phenyl substituted by halogen; CrC6 alkoxy; Ci-C6 alkyl optionally substituted by halogen or hydroxy; or C3-C6 cycloalkyl optionally substituted by halogen, hydroxy, or C C6 alkyl optionally substituted by halogen; X stands for O and R10 is absent; or X stands for N, and R10 is hydrogen or -CO-R1 1 , and the remaining variables are as defined in claim 1.
5. A compound according to claim 1 , or a pharmaceutically acceptable salt thereof, wherein R1 is hydrogen, methyl or hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 is hydrogen; R5 is phenyl substituted by CrC6 alkoxy; CrC6 alkyl optionally substituted by halogen or hydroxy; or C3-C6 cycloalkyi optionally substituted by halogen, hydroxy, or C C6 alkyl optionally substituted by halogen; X stands for N, R10 is hydrogen or -CO-R1 1 , R1 1 stands for NR6R7 wherein R6 and R7 are independently hydrogen or methyl; and the remaining variables are as defined in claim 1.
6. A compound according to claim 1 , or a pharmaceutically acceptable salt thereof, wherein R1 is hydrogen, methyl or hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 is hydrogen, R5 is azetidine optionally substituted by CrC6 alkoxy, X stands for N, and R10 is hydrogen or -CO-R1 1 , and the remaining variables are as defined in claim 1.
7. A compound according to claim 1 , or a pharmaceutically acceptable salt thereof, wherein R1 is hydrogen, methyl or hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one optionally substituted by C3-C6 cycloalkyi or CrC6 alkyl optionally substituted by hydroxy, X stands for N, and R10 is hydrogen or -CO-R1 1 , and the remaining variables are as defined in claim 1.
8. A compound according to claim 1 , or a pharmaceutically acceptable salt thereof, wherein R1 is hydrogen, methyl or hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one substituted by C3-C6 cycloalkyi or CrC6 alkyl optionally substituted by hydroxy in the 6- position of said isoquinolin-ring, X stands for N, and R10 is hydrogen or -CO-R1 1 , and the remaining variables are as defined in claim 1.
9. A compound according to claim 1 , or a pharmaceutically acceptable salt thereof, wherein R1 is hydrogen, methyl or hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 is hydrogen, R5 is azetidine optionally substituted by CrC6 alkoxy, X stands for O, and the remaining variables are as defined in claim 1.
10. A compound according to claim 1 , or a pharmaceutically acceptable salt thereof, wherein R1 is hydrogen, methyl or hydroxymethyl , R2 and R3 are independently hydrogen or fluoro, R4 together with R5 is a 3,4-dihydro-2H-isoquinolin-1-one optionally substituted by C3-C6 cycloalkyi or CrC6 alkyl optionally substituted by hydroxy, X stands for O, and the remaining variables are as defined claim 1.
11. A compound of claim 1 , or a pharmaceutically acceptable salt thereof, which is selected from:
4-(4-{5-Fluoro-3-[4-(1-fluoro-cyclopropyl)-benzoylamino]-2-methyl-phenyl}-7H- pyrrolo[2'3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 4-(4-{3-[(3,3-Dimethyl-2,3-dihydro-benzofuran-6-carbonyl)-amino]-5-fluoro-2-methyl- phenyl}-7H-pyrrolo[2'3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-(4-{5-Fluoro-2-methyl-3-[(5-methyl-4,5,6,7-tetrahydro-benzo[b]thiophene-2-carbonyl)- amino]-phenyl}-7H-pyrrolo[2'3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-(4-{5-Fluoro-3-[4-isopropyl-methyl-amino)-benzoylamino]-2-methyl-phenyl}-7H- pyrrolo[2'3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
3- Methyl-1 H-indole-6-carboxylic acid {3-[6-(1-dimethylcarbamoyl-1 ,2,3,6-tetrahydro- pyridin-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5-fluoro-2-methyl-phenyl}-amide,
4- (4-{5-Fluoro-3-[4-(2-hydroxy-1 ,1-dimethyl-ethyl)-benzoylamino]-2-methyl-phenyl}-7H- pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 4-{4-[5-Fluoro-2-methyl-3-(4-piperidin-1-yl-benzoylamino)-phenyl]-7H-pyrrolo[2,3- d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-{4-[5-Fluoro-3-(isopropenyl-benzoylamino)-2-methyl-phenyl]-7H-pyrrolo[2,3- d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-(4-{5-Fluoro-2-methyl-3-[4-(1-trifluoromethyl-cyclopropyl)-benzoylamino]-phenyl}-7H- pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 4-{4-[5-Fluoro-3-(4-isopropoxy-benzoylamino)-2-methyl-phenyl]-7H-pyrrolo[2,3- d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 4-(4-{5-Fluoro-3-[4-pentafluorothio-benzoylamino]-2-methyl-phenyl}-7H-pyrrolo[2,3- d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-(4-{5-Fluoro-3-[4-(2-methoxy-1 ,1 -dimethyl-ethyl )-benzoylamino]-2-methyl-phenyl}-7H- pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 4-(4-{5-Fluoro-3-[4-(1-methoxy-1 -methyl-ethyl )-benzoylamino]-2-methyl-phenyl}-7H- pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 1-Methyl-1 H-pyrrolo[2,3-b]pyridine-6-carboxylic acid {3-[6-(1-dimethyl-carbamoyl-1 ,2,3,6- tetrahydro-pyridin-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5-fluoro-2-methyl-phenyl}-amide, 4-{4-[3-(4-Dimethylamino-benzoylamino)-5-fluoro-2-methyl-phenyl]-7H-pyrrolo[2,3- d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-(4-{3-[2-Fluoro-4-(1-hydroxy-1 -methyl-ethyl )-benzoylamino]-2-hydroxymethyl-phenyl}- 7H-pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid
dimethylamide,
4-{4-[3-(4-Cyclopropyl-benzoylamino)-2-hydroxymethyl-phenyl]-7H-pyrrolo[2,3- d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-(4-{5-Fluoro-2-methyl-3-[4-(2,2,2-trifluoro-1-hydroxy-1 -methyl-ethyl )-benzoylamino]- phenyl}-7H-pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-{4-[3-(4-Acetyl-benzoylamino)-5-fluoro-2-methyl-phenyl]-7H-pyrrolo[2,3-d]pyrimidin-6- yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-{4-[3-(4-Cyclopropyl-benzoylamino)-4-fluoro-phenyl]-7H-pyrrolo[2,3-d]pyrimidin-6-yl}- 3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-(4-{4-Fluoro-3-[4-(2-hydroxy-1 , 1-dimethyl-ethyl)-benzoylamino]-phenyl}-7H-pyrrolo[2,3- d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
N-{3-[6-(3,6-Dihydro-2H-pyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5-fluoro-2-methyl- phenyl}-2-fluoro-4-(1-hydroxy-1 -methyl-ethyl )-benzamide,
N-{3-[6-(3,6-Dihydro-2H-pyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2-hydroxymethyl- phenyl}-2-fluoro-4-(1-hydroxy-1 -methyl-ethyl )-benzamide,
4-tert-Butyl-N-{3-[6-(3,6-dihydro-2H-thiopyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5- fluoro-2-methyl-phenyl}-benzamide,
4-tert-Butyl-N-{5-fluoro-2-methyl-3-[6-(3,6-dihydro-1-oxido-2H-thiopyran-4-yl)-7H- pyrrolo[2,3-d]pyrimidin-4-yl]-phenyl}-benzamide,
4-tert-Butyl-N-{3-[6-(3,6-dihydro-1 ,1-dioxido-2H-thiopyran-4-yl)-7H-pyrrolo[2,3- d]pyrimidin-4-yl]-5-fluoro-2-methyl-phenyl}-benzamide, 4-tert-Butyl-N-{3-[6-(1 ^-dioxa-spiro[4.5]dec-7-en-8-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5- fluoro-2-methyl-phenyl}-benzamide,
4 ert-Butyl-N-{5-fluoro-3-[6-(4-hydroxy-cyclohex-1-enyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]- 2-methyl-phenyl}-benzamide,
4-{4-[3-(4-Cyclopropyl-benzoylamino)-5-fluoro-2-methyl-phenyl]-7H-pyrrolo[2'3- d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-Cyclopropyl-N-(5-fluoro-2-methyl-3-{6-[1-(pyrrolidine-1-carbonyl)-1 ,2,3,6-tetrahydro- pyridin-4-yl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}-phenyl)-benzamide,
Acetic acid 2-(4-{4-[3-(4-cyclopropyl-benzoylamino)-5-fluoro-2-methyl-phenyl]-7H- pyrrolo[2,3-d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridin-1-yl)-2-oxo-ethyl ester,
4-Cyclopropyl-N-(5-fluoro-3-{6-[1-(2-hydroxy-actyl)-1 ,2,3,6-tetrahydro-pyridin-4-yl]-7H- pyrrolo[2,3-d]pyrimidin-4-yl}-2-methyl-phenyl)benzamide,
N-(3-{6-[1-(2-Cyano-acetyl)-1 ,2,3,6 etrahydro-pyridin-4-yl]-7H-pyrrolo[2,3-d]pyrimidin^ yl}-5-fluoro-2-methyl-phenyl)-4-cyclopropyl-benzamide,
N-(5-Fluoro-2-methyl-3-{6-[1-(pyrrolidine-1-carbo
pyrrolo[2,3-d]pyrimidin-4-yl}-phenyl)-4-(pentafluoro-sulfanyl)-benzamide,
Acetic acid 2-[4-(4-{5-fluoro-2-methyl-3-[4-(pentafluoro-sulfanyl)-benzoylamino]-phenyl}-
7H-pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridin-1-yl]-2-oxo-ethyl ester,
N-(5-Fluoro-3-{6-[1-(2-hydroxy-acetyl)-1 ,2,3,6-tetrahydro-pyridin-4-yl]-7H-pyrrolo[2,3- d]pyrimidin-4-yl}-2-methyl-phenyl)-4-(pentafluoro-sulfanyl)-benzamide,
4-{4-[3-(4 ert-Butyl-benzoylamino)-2-(tert-butyl-diphenyl-silanyloxymethyl)-phenyl]-7H pyrrolo[2,3-d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester,
4-tert-Butyl-N-(3-{6-[1-(2-fluoro-acetyl)-1 ,2,3,6-tetrahydro-pyridin-4-yl]-7H-pyrrolo[2,3- d]pyrimidin-4-yl}-2-hydroxymethyl-phenyl)-benzamide,
4-(4-{5-Fluoro-3-[2-fluoro-4-(1-hydroxy-1 -methyl-ethyl )-benzoylamino]-2-methyl-phenyl}- 7H-pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid
dimethylamide,
4-tert-Butyl-N-{5-fluoro-2-methyl-3-[6-(1 ,2,3,6-tetrahydro-pyridin-4-yl)-7H-pyrrolo[2,3- d]pyrimidin-4-yl]-phenyl}-benzamide,
4-{4-[3-(4-tert-Butyl-benzoylamino)-5-fluoro-2-methyl-phenyl]-7H-pyrrolo[2,3-d]pyrimidin- 6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-tert-Butyl-N-{5-fluoro-3-[6-(1-methanesulfonyl-1 ,2,3,6-tetrahydro-pyridin-4-yl)-7H- pyrrolo[2,3-d]pyrimidin-4-yl]-2-methyl-phenyl}-benzamide, 4-tert-Butyl-N-{3-[6-(1-dimethylsulfamoyl-1 ,2,3,6-tetrahydro-pyridin-4-yl)-7H-pyrrolo[2,3 d]pyrimidin-4-yl]-5-fluoro-2-methyl-phenyl}-benzamide,
4-tert-Butyl-N-{3-[6-(1-methanesulfonyl-1 ,2,3,6-tetrahydro-pyridin-4-yl)-7H-pyrrolo[2,3- d]pyrimidin-4-yl]-2-methyl-phenyl}-benzamide,
4-{4-[3-(6 ert-Butyl-1-oxo-3,4-dihydro-1 H-isoquinolin-2-yl)-2-hydroxymethyl-phenyl]-7H pyrrolo[2,3-d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 4-{4-[3-(6-Cyclopropyl-1-oxo-3,4-dihydro-1 H-isoquinolin-2-yl)-2-hydroxymethyl-phenyl]- 7H-pyrrolo[2,3-d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid
dimethylamide,
4-(4-{2-Hydroxymethyl-3-[6-(1-hydroxy-1 -methyl-ethyl )-1-oxo-3, 4-dihydro-1 H-isoquinolin- 2-yl]-phenyl}-7H-pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-tert-Butyl-N-{3-[6-(3,6-dihydro-2H-pyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2- hydroxymethyl-phenyl}-benzamide,
4-tert-Butyl-N-{3-[6-(3,6-dihydro-2H-pyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5-fluoro- 2-methyl-phenyl}-benzamide,
N-{3-[6-(3,6-Dihydro-2H-pyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5-fluoro-2-methyl- phenyl}-4-dimethylamino-benzamide,
4-tert-Butyl-N-{3-[6-(3,6-dihydro-2H-thiopyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2- methyl-phenyl}-benzamide,
4-tert-Butyl -N-{2-methyl-3-[6-(3,6-dihydro-1-oxido-2H-thiopyran-4-yl)-7H-pyrrolo[2,3- d]pyrimidin-4-yl]-phenyl}-benzamide,
4-tert-Butyl -N-{3-[6-(3,6-dihydro-1 ,1-dioxido-2H-thiopyran-4-yl)-7H-pyrrolo[2,3- d]pyrimidin-4-yl]-2-methyl-phenyl}-benzamide,
4-tert-Butyl-N-{3-[6-(1 ,4-dioxa-spiro[4.5]dec-7-en-8-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2- hydroxymethyl-phenyl}-benzamide,
4-tert-Butyl-N-{3-[6-(4-hydroxy-cyclohex-1-enyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2- hydroxymethyl-phenyl}-benzamide,
4-tert-Butyl-N-{3-[6-(4-dimethylamino-cyclohex-1-enyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2- hydroxymethyl-phenyl}-benzamide,
4-(4-{3-[(3-tert-Butoxy-azetidine-1-carbonyl)-amino]-5-fluoro-2-methyl-phenyl}-7H- pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 4-(4-{5-Fluoro-3-[(3-isopropoxy-azetidine-1-carbonyl)-amino]-2-methyl-phenyl}-7H- pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 5-Fluoro-1 ,3-dihydro-isoindole-2-carboxylic acid {3-[6-(1-dimethylcarbamoyl-1 ,2,3,6- tetrahydro-pyridin-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5-fluoro-2-methyl-phenyl}-am 4-[4-(5-Fluoro-2-methyl-3-{[3-(2,2,2-trifluoro-1-trifluoromethyl-ethoxy)-azetidine-1- carbonyl]-amino}-phenyl)-7H-pyrrolo[2,3-d]pyrimidin-6-yl]-3,6-dihydro-2H-pyridine-1- carboxylic acid dimethylamide,
3- tert-Butoxy-azetidine-1-carboxylic acid {3-[6-(3,6-dihydro-2H-pyran-4-yl)-7H- pyrrolo[2,3-d]pyrimidin-4-yl]-5-fluoro-2-methyl-phenyl}-amide,
4- (4-{3-[(3-tert-Butoxy-azetidine-1-carbonyl)-amino]-4-fluoro-phenyl}-7H-pyrrolo[2,3- d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide,
4-(4-{3-[(3-tert-Butoxy-azetidine-1-carbonyl)-amino]-4-fluoro-2-methyl-phenyl}-7H- pyrrolo[2,3-d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide, 4-tert-Butyl-N-{3-[6-(3,6-dihydro-2H-pyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2- methyl-phenyl}-benzamide,
4 ert-Butyl-N-{3-[6-(1 ,4-dioxa-spiro[4.5]dec-7-en-8-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2- methyl-phenyl}-benzamide,
4-tert-Butyl-N-{3-[6-(4-hydroxy-cyclohex-1-enyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2- methyl-phenyl}-benzamide, and
4-tert-Butyl-N-{3-[6-(4-dimethylamino-cyclohex-1-enyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2- methyl-phenyl}-benzamide.
12. A compound claim 1 , or a pharmaceutically acceptable salt thereof, which is selected from:
4-{4-[3-(4-Acetyl-benzoylamino)-5-fluoro-2-methyl-phenyl]-7H-pyrrolo[2,3-d]pyrimidin-6- yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide;
4-{4-[3-(4-Cyclopropyl-benzoylamino)-4-fluoro-phenyl]-7H-pyrrolo[2,3-d]pyrimidin-6-yl}- 3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide;
4-(4-{4-Fluoro-3-[4-(2-hydroxy-1 , 1-dimethyl-ethyl)-benzoylamino]-phenyl}-7H-pyrrolo[2,3- d]pyrimidin-6-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide;
N-{3-[6-(3,6-Dihydro-2H-pyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5-fluoro-2-methyl- phenyl}-2-fluoro-4-(1-hydroxy-1 -methyl-ethyl )-benzamide;
N-{3-[6-(3,6-Dihydro-2H-pyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-2-hydroxymethyl- phenyl}-2-fluoro-4-(1-hydroxy-1 -methyl-ethyl )-benzamide;
4-tert-Butyl-N-{3-[6-(3,6-dihydro-2H-thiopyran-4-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5- fluoro-2-methyl-phenyl}-benzamide; 4-tert-Butyl-N-{5-fluoro-2-methyl-3-[6-(3,6-dihydro-1-oxido-2H-thiopyran-4-yl)-7H- pyrrolo[2,3-d]pyrimidin-4-yl]-phenyl}-benzamide;
4-tert-Butyl-N-{3-[6-(3,6-dihydro-1 ,1-dioxido-2H-thiopyran-4-yl)-7H-pyrrolo[2,3- d]pyrimidin-4-yl]-5-fluoro-2-methyl-phenyl}-benzamide;
4-tert-Butyl-N-{3-[6-(1 ^-dioxa-spiro[4.5]dec-7-en-8-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]-5- fluoro-2-methyl-phenyl}-benzamide;
4 ert-Butyl-N-{5-fluoro-3-[6-(4-hydroxy-cyclohex-1-enyl)-7H-pyrrolo[2,3-d]pyrimidin-4-yl]^ 2-methyl-phenyl}-benzamide;
4-{4-[3-(4-Cyclopropyl-benzoylamino)-5-fluoro-2-methyl-phenyl]-7H-pyrrolo[2'3- d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridine-1-carboxylic acid dimethylamide;
4-Cyclopropyl-N-(5-fluoro-2-methyl-3-{6-[1-(pyrrolidine-1-carbonyl)-1 ,2,3,6-tetrahydro- pyridin-4-yl]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}-phenyl)-benzamide;
Acetic acid 2-(4-{4-[3-(4-cyclopropyl-benzoylamino)-5-fluoro-2-methyl-phenyl]-7H- pyrrolo[2,3-d]pyrimidin-6-yl}-3,6-dihydro-2H-pyridin-1-yl)-2-oxo-ethyl ester; and
4-Cyclopropyl-N-(5-fluoro-3-{6-[1-(2-hydroxy-actyl)-1 ,2,3,6-tetrahydro-pyridin-4-yl]-7H- pyrrolo[2,3-d]pyrimidin-4-yl}-2-methyl-phenyl)benzamide.
13. A pharmaceutical composition comprising a therapeutically effective amount of a compound according to any one of claims 1 to 12 and one or more pharmaceutically acceptable carriers.
14. A combination comprising a therapeutically effective amount of a compound according to any one of claims 1 to 12 or a pharmaceutically acceptable salt thereof and one or more therapeutically active co-agents.
15. A method of modulating Btk activity in a subject, wherein the method comprises administering to the subject a therapeutically effective amount of the compound according to any one of claims 1 to 12 or a pharmaceutically acceptable salt thereof.
16. A compound according to any one of claims 1 to 12 or a pharmaceutically acceptable salt thereof, for use as a medicament.
17. Use of a compound according to any one of claims 1 to 12 or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a disorder or disease mediated by Btk.
PCT/IB2012/001699 2011-07-08 2012-07-07 Novel pyrrolo pyrimidine derivatives WO2013008095A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
ES12761655.5T ES2548414T3 (en) 2011-07-08 2012-07-07 Novel pyrimidine pyrrolo derivatives
EA201490229A EA201490229A1 (en) 2011-07-08 2012-07-07 NEW PYRROPHYRIMIDINE DERIVATIVES
JP2014517980A JP6145451B2 (en) 2011-07-08 2012-07-07 Novel pyrrolopyrimidine derivatives
KR1020147002986A KR20140058543A (en) 2011-07-08 2012-07-07 Novel pyrrolo pyrimidine derivatives
EP12761655.5A EP2729466B1 (en) 2011-07-08 2012-07-07 Novel pyrrolo pyrimidine derivatives
AU2012282229A AU2012282229B2 (en) 2011-07-08 2012-07-07 Novel pyrrolo pyrimidine derivatives
CN201280033935.9A CN103732596B (en) 2011-07-08 2012-07-07 Pyrrolopyrimidine derivatives
MX2014000338A MX2014000338A (en) 2011-07-08 2012-07-07 Novel pyrrolo pyrimidine derivatives.
CA2841111A CA2841111A1 (en) 2011-07-08 2012-07-07 Novel pyrrolo pyrimidine derivatives
US14/130,536 US9233111B2 (en) 2011-07-08 2012-07-07 Pyrrolo pyrimidine derivatives
BR112014000314A BR112014000314A2 (en) 2011-07-08 2012-07-07 pyrrole pyrimidine derivatives

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161505560P 2011-07-08 2011-07-08
US61/505,560 2011-07-08

Publications (1)

Publication Number Publication Date
WO2013008095A1 true WO2013008095A1 (en) 2013-01-17

Family

ID=46880751

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2012/001699 WO2013008095A1 (en) 2011-07-08 2012-07-07 Novel pyrrolo pyrimidine derivatives

Country Status (12)

Country Link
US (1) US9233111B2 (en)
EP (1) EP2729466B1 (en)
JP (1) JP6145451B2 (en)
KR (1) KR20140058543A (en)
CN (1) CN103732596B (en)
AU (1) AU2012282229B2 (en)
BR (1) BR112014000314A2 (en)
CA (1) CA2841111A1 (en)
EA (1) EA201490229A1 (en)
ES (1) ES2548414T3 (en)
MX (1) MX2014000338A (en)
WO (1) WO2013008095A1 (en)

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013157022A1 (en) 2012-04-20 2013-10-24 Advinus Therapeutics Limited Substituted hetero-bicyclic compounds, compositions and medicinal applications thereof
WO2013157021A1 (en) 2012-04-20 2013-10-24 Advinus Therapeutics Limited Bicyclic compounds, compositions and medicinal applications thereof
US9233111B2 (en) 2011-07-08 2016-01-12 Novartis Ag Pyrrolo pyrimidine derivatives
EP3042903A1 (en) 2015-01-06 2016-07-13 Advinus Therapeutics Limited Substituted hetero-bicyclic compounds, compositions and medicinal applications thereof
US9512084B2 (en) 2013-11-29 2016-12-06 Novartis Ag Amino pyrimidine derivatives
WO2017106634A1 (en) * 2015-12-17 2017-06-22 Incyte Corporation N-phenyl-pyridine-2-carboxamide derivatives and their use as pd-1/pd-l1 protein/protein interaction modulators
WO2018053437A1 (en) 2016-09-19 2018-03-22 Mei Pharma, Inc. Combination therapy
US10308644B2 (en) 2016-12-22 2019-06-04 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2019186358A1 (en) 2018-03-26 2019-10-03 Novartis Ag 3-hydroxy-n-(3-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)phenyl)pyrrolidine-1-carboxamide derivatives
WO2019186343A1 (en) 2018-03-26 2019-10-03 Novartis Ag N-(3-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)phenyl)benzamide derivatives
EP3550031A1 (en) 2012-07-24 2019-10-09 Pharmacyclics, LLC Mutations associated with resistance to inhibitors of bruton's tyrosine kinase (btk)
EP3556761A4 (en) * 2017-02-08 2019-12-25 The National Institutes Of Pharmaceutical Research Pyrrolo-aromatic heterocyclic compound, preparation method therefor, and medical use thereof
US10618916B2 (en) 2018-05-11 2020-04-14 Incyte Corporation Heterocyclic compounds as immunomodulators
US10669271B2 (en) 2018-03-30 2020-06-02 Incyte Corporation Heterocyclic compounds as immunomodulators
JP2020105181A (en) * 2013-12-02 2020-07-09 ファーマサイクリックス エルエルシー Methods of treating and preventing alloantibody driven chronic graft versus host disease
US10793565B2 (en) 2016-12-22 2020-10-06 Incyte Corporation Heterocyclic compounds as immunomodulators
US10806785B2 (en) 2016-12-22 2020-10-20 Incyte Corporation Immunomodulator compounds and methods of use
US11401279B2 (en) 2019-09-30 2022-08-02 Incyte Corporation Pyrido[3,2-d]pyrimidine compounds as immunomodulators
US11407749B2 (en) 2015-10-19 2022-08-09 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2022189859A1 (en) 2021-03-12 2022-09-15 Novartis Ag Fatty acid-bifunctional degrader conjugates and their methods of use
US11465981B2 (en) 2016-12-22 2022-10-11 Incyte Corporation Heterocyclic compounds as immunomodulators
US11535615B2 (en) 2015-12-22 2022-12-27 Incyte Corporation Heterocyclic compounds as immunomodulators
US11572366B2 (en) 2015-11-19 2023-02-07 Incyte Corporation Heterocyclic compounds as immunomodulators
US11608337B2 (en) 2016-05-06 2023-03-21 Incyte Corporation Heterocyclic compounds as immunomodulators
US11613536B2 (en) 2016-08-29 2023-03-28 Incyte Corporation Heterocyclic compounds as immunomodulators
US11623921B2 (en) 2014-10-24 2023-04-11 Bristol-Myers Squibb Company Indole carboxamide compounds
US11673883B2 (en) 2016-05-26 2023-06-13 Incyte Corporation Heterocyclic compounds as immunomodulators
US11718605B2 (en) 2016-07-14 2023-08-08 Incyte Corporation Heterocyclic compounds as immunomodulators
US11753406B2 (en) 2019-08-09 2023-09-12 Incyte Corporation Salts of a PD-1/PD-L1 inhibitor
US11760756B2 (en) 2020-11-06 2023-09-19 Incyte Corporation Crystalline form of a PD-1/PD-L1 inhibitor
US11780836B2 (en) 2020-11-06 2023-10-10 Incyte Corporation Process of preparing a PD-1/PD-L1 inhibitor
US11866434B2 (en) 2020-11-06 2024-01-09 Incyte Corporation Process for making a PD-1/PD-L1 inhibitor and salts and crystalline forms thereof
US11866451B2 (en) 2019-11-11 2024-01-09 Incyte Corporation Salts and crystalline forms of a PD-1/PD-L1 inhibitor
US11873309B2 (en) 2016-06-20 2024-01-16 Incyte Corporation Heterocyclic compounds as immunomodulators

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ711540A (en) 2013-04-25 2018-08-31 Beigene Ltd Fused heterocyclic compounds as protein kinase inhibitors
EP4130044A1 (en) 2013-09-13 2023-02-08 BeiGene Switzerland GmbH Anti-pd1 antibodies and their use as therapeutics and diagnostics
TWI726608B (en) 2014-07-03 2021-05-01 英屬開曼群島商百濟神州有限公司 Anti-pd-l1 antibodies and their use as therapeutics and diagnostics
NZ729603A (en) * 2014-10-06 2022-02-25 Merck Patent Gmbh Heteroaryl compounds as btk inhibitors and uses thereof
WO2016079669A1 (en) * 2014-11-19 2016-05-26 Novartis Ag Labeled amino pyrimidine derivatives
AR102871A1 (en) * 2014-12-03 2017-03-29 Pharmacyclics Llc FIBROSIS TREATMENT METHODS
AU2017293423B2 (en) 2016-07-05 2023-05-25 Beigene, Ltd. Combination of a PD-1 antagonist and a RAF inhibitor for treating cancer
CN109563099B (en) 2016-08-16 2023-02-03 百济神州有限公司 Crystal form of compound, preparation and application thereof
WO2018033135A1 (en) 2016-08-19 2018-02-22 Beigene, Ltd. Use of a combination comprising a btk inhibitor for treating cancers
US11555038B2 (en) 2017-01-25 2023-01-17 Beigene, Ltd. Crystalline forms of (S)-7-(1-(but-2-ynoyl)piperidin-4-yl)-2-(4-phenoxyphenyl)-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide, preparation, and uses thereof
AU2018290532A1 (en) 2017-06-26 2019-11-21 Beigene, Ltd. Immunotherapy for hepatocellular carcinoma
CN110997677A (en) 2017-08-12 2020-04-10 百济神州有限公司 Btk inhibitors with improved dual selectivity
WO2019108795A1 (en) 2017-11-29 2019-06-06 Beigene Switzerland Gmbh Treatment of indolent or aggressive b-cell lymphomas using a combination comprising btk inhibitors
CN111704617B (en) * 2020-06-15 2022-08-23 嘉兴特科罗生物科技有限公司 Small molecule compound
CN115073469B (en) * 2021-03-15 2023-12-22 药雅科技(上海)有限公司 Preparation and application of pyrrolopyrimidine compound as kinase inhibitor
US11786531B1 (en) 2022-06-08 2023-10-17 Beigene Switzerland Gmbh Methods of treating B-cell proliferative disorder

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002038561A1 (en) 2000-11-07 2002-05-16 Novartis Ag Indolylmaleimide derivatives as protein kinase c inhibitors
WO2003082859A1 (en) 2002-04-03 2003-10-09 Novartis Ag Indolylmaleimide derivatives
WO2004052359A1 (en) 2002-12-09 2004-06-24 The Board Of Regents Of The University Of Texas System Methods for selectively inhibiting janus tyrosine kinase 3 (jak3)
WO2004078163A2 (en) 2003-02-28 2004-09-16 Transform Pharmaceuticals, Inc. Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
WO2005000833A1 (en) 2003-05-19 2005-01-06 Irm, Llc Immunosuppressant compounds and compositions
WO2005066156A1 (en) 2004-01-12 2005-07-21 Cytopia Research Pty Ltd Selective kinase inhibitors
WO2007090752A1 (en) 2006-02-07 2007-08-16 F. Hoffmann-La Roche Ag Anthranilamide/2-amino-heteroarene carboxamide derivatives
WO2007145834A2 (en) 2006-06-08 2007-12-21 Amgen Inc. Benzamide derivatives and uses related thereto
US20090105209A1 (en) 2007-10-23 2009-04-23 Roche Palo Alto Llc BTK protein kinase inhibitors
WO2009077334A1 (en) 2007-12-14 2009-06-25 F. Hoffmann-La Roche Ag Novel imidazo[1,2-a]pyridine and imidazo[1,2-b]pyridazine derivatives
US20090306041A1 (en) 2008-02-05 2009-12-10 Roche Palo Alto Llc Inhibitors of Bruton's tyrosine kinase
WO2010000633A1 (en) 2008-07-02 2010-01-07 F. Hoffmann-La Roche Ag Novel phenylpyrazinones as kinase inhibitors

Family Cites Families (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6303652B1 (en) 1998-08-21 2001-10-16 Hughes Institute BTK inhibitors and methods for their identification and use
SK18542000A3 (en) 1998-06-04 2001-12-03 Abbott Laboratories Cell adhesion-inhibiting anti-inflammatory compounds
PA8474101A1 (en) 1998-06-19 2000-09-29 Pfizer Prod Inc PYROLEUM [2,3-D] PIRIMIDINE COMPOUNDS
GB2345868A (en) 1999-01-23 2000-07-26 Martin Hodgson Self adhesive magnetic or electromagnetic tapes
WO2000075145A1 (en) 1999-06-03 2000-12-14 Abbott Laboratories Cell adhesion-inhibiting antiinflammatory compounds
GB0005345D0 (en) 2000-03-06 2000-04-26 Mathilda & Terence Kennedy Ins Methods of treating sepsis septic shock and inflammation
WO2002038797A2 (en) 2000-10-23 2002-05-16 Bristol-Myers Squibb Company Modulators of bruton's tyrosine kinase, their identification and use
EP1217000A1 (en) 2000-12-23 2002-06-26 Aventis Pharma Deutschland GmbH Inhibitors of factor Xa and factor VIIa
GB0119249D0 (en) * 2001-08-07 2001-10-03 Novartis Ag Organic compounds
EP2322201A3 (en) 2002-10-29 2011-07-27 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
EP1473039A1 (en) 2003-05-02 2004-11-03 Centre National De La Recherche Scientifique (Cnrs) Use of inhibitors and antisense oligonucleotides of BTK for the treatment of proliferative mastocytosis
US7393848B2 (en) 2003-06-30 2008-07-01 Cgi Pharmaceuticals, Inc. Certain heterocyclic substituted imidazo[1,2-A]pyrazin-8-ylamines and methods of inhibition of Bruton's tyrosine kinase by such compounds
PE20051092A1 (en) * 2004-01-29 2006-01-20 Novartis Ag PYROLO-PYRIMIDINE DERIVATIVES USEFUL FOR THE TREATMENT OF PROLIFERATIVE DISEASES
JP2008508358A (en) 2004-08-02 2008-03-21 オーエスアイ・ファーマスーティカルズ・インコーポレーテッド Aryl-amino substituted pyrrolopyrimidine multikinase inhibitor compounds
GB0420719D0 (en) 2004-09-17 2004-10-20 Addex Pharmaceuticals Sa Novel allosteric modulators
WO2006099075A2 (en) 2005-03-10 2006-09-21 Cgi Pharmaceuticals, Inc. Certain substituted amides, method of making, and method of use thereof
WO2006101783A2 (en) 2005-03-15 2006-09-28 Irm Llc Compounds and compositions as protein kinase inhibitors
MX2007014066A (en) 2005-05-13 2008-02-11 Irm Llc Compounds and compositions as protein kinase inhibitors.
CA2608726C (en) 2005-05-20 2013-07-09 Methylgene Inc. Inhibitors of vegf receptor and hgf receptor signaling
EP1963320A1 (en) * 2005-12-07 2008-09-03 OSI Pharmaceuticals, Inc. Pyrrolopyridine kinase inhibiting compounds
TW201434835A (en) 2005-12-13 2014-09-16 Incyte Corp Heteroaryl substituted pyrrolo[2,3-b]pyridines and pyrrolo[2,3-b]pyrimidines as janus kinase inhibitors
US7915411B2 (en) 2005-12-21 2011-03-29 Abbott Laboratories Anti-viral compounds
US20070208053A1 (en) 2006-01-19 2007-09-06 Arnold Lee D Fused heterobicyclic kinase inhibitors
TWI398252B (en) 2006-05-26 2013-06-11 Novartis Ag Pyrrolopyrimidine compounds and their uses
DE102006033109A1 (en) 2006-07-18 2008-01-31 Grünenthal GmbH Substituted heteroaryl derivatives
US20100160292A1 (en) 2006-09-11 2010-06-24 Cgi Pharmaceuticals, Inc Kinase Inhibitors, and Methods of Using and Identifying Kinase Inhibitors
AR063946A1 (en) 2006-09-11 2009-03-04 Cgi Pharmaceuticals Inc CERTAIN REPLACED PIRIMIDINS, THE USE OF THE SAME FOR THE TREATMENT OF DISEASES MEDIATED BY THE INHIBITION OF THE ACTIVITY OF BTK AND PHARMACEUTICAL COMPOSITIONS THAT UNDERSTAND THEM.
HUE028086T2 (en) 2006-09-22 2016-11-28 Pharmacyclics Llc Inhibitors of bruton's tyrosine kinase
DK2134374T3 (en) 2007-03-14 2014-02-24 Bionsil S R L In Liquidazione BTK-INHIBITORS FOR USE IN PROCESSING chemotherapeutic agent RESISTANT TUMORS Epithelial
CA3001152A1 (en) 2007-03-28 2008-10-09 Pharmacyclics Llc Inhibitors of bruton's tyrosine kinase
WO2008144253A1 (en) 2007-05-14 2008-11-27 Irm Llc Protein kinase inhibitors and methods for using thereof
MX2010000617A (en) 2007-07-17 2010-05-17 Plexxikon Inc Compounds and methods for kinase modulation, and indications therefor.
US20090118276A1 (en) * 2007-11-02 2009-05-07 Wyeth Thienopyrimidines, thienopyridines, and pyrrolopyrimidines as b-raf inhibitors
PE20091846A1 (en) 2008-05-19 2009-12-16 Plexxikon Inc PIRROLO [2,3-d] -PYRIMIDINE DERIVATIVES AS KINE MODULATORS
WO2009143018A2 (en) 2008-05-19 2009-11-26 Plexxikon, Inc. Compounds and methods for kinase modulation, and indications therefor
WO2010009342A2 (en) 2008-07-16 2010-01-21 Pharmacyclics, Inc. Inhibitors of bruton's tyrosine kinase for the treatment of solid tumors
JP2012502104A (en) 2008-09-10 2012-01-26 カリプシス・インコーポレーテッド Aminopyrimidine inhibitors against histamine receptors for the treatment of disease
WO2010036316A1 (en) 2008-09-24 2010-04-01 Yangbo Feng Urea and carbamate compounds and analogs as kinase inhibitors
JP5731978B2 (en) 2008-09-26 2015-06-10 インテリカイン, エルエルシー Heterocyclic kinase inhibitor
CL2009001884A1 (en) 2008-10-02 2010-05-14 Incyte Holdings Corp Use of 3-cyclopentyl-3- [4- (7h-pyrrolo [2,3-d] pyrimidin-4-yl) -1h-pyrazol-1-yl) propanonitrile, janus kinase inhibitor, and use of a composition that understands it for the treatment of dry eye.
US20100261776A1 (en) 2008-11-07 2010-10-14 The Research Foundation Of State University Of New York Bruton's tyrosine kinase as anti-cancer drug target
WO2010068806A1 (en) 2008-12-10 2010-06-17 Cgi Pharmaceuticals, Inc. Amide derivatives as btk inhibitors in the treatment of allergic, autoimmune and inflammatory disorders as well as cancer
EP2416772A1 (en) 2009-04-06 2012-02-15 PTC Therapeutics, Inc. Combinations of a hcv inhibitor such as bicyclic pyrrole derivatives and a therapeutic agent
WO2010117935A1 (en) 2009-04-06 2010-10-14 Schering Corporation Compounds and methods for antiviral treatment
WO2010129053A2 (en) 2009-05-05 2010-11-11 Dana Farber Cancer Institute Egfr inhibitors and methods of treating disorders
CA2771822C (en) 2009-09-04 2020-08-11 Daniel A. Erlanson Bruton's tyrosine kinase inhibitors
WO2011029043A1 (en) 2009-09-04 2011-03-10 Biogen Idec Ma Inc. Heteroaryl btk inhibitors
MX2012013196A (en) 2010-05-12 2013-03-22 Abbvie Inc Pyrrolopyridine and pyrrolopyrimidine inhibitors of kinases.
US20120071497A1 (en) 2010-06-03 2012-03-22 Pharmacyclics, Inc. Methods of treating abc-dlbcl using inhibitors of bruton's tyrosine kinase
WO2012058645A1 (en) 2010-10-29 2012-05-03 Biogen Idec Ma Inc. Heterocyclic tyrosine kinase inhibitors
MX2014000130A (en) 2011-06-28 2014-05-01 Pharmacyclics Inc Methods and compositions for inhibition of bone resorption.
KR20140058543A (en) 2011-07-08 2014-05-14 노파르티스 아게 Novel pyrrolo pyrimidine derivatives
EP3778896A1 (en) 2011-08-09 2021-02-17 Fred Hutchinson Cancer Research Center Methods and compositions for inhibiting the growth and/or proliferation of myc-driven tumor cells
DE102011111400A1 (en) 2011-08-23 2013-02-28 Merck Patent Gmbh Bicyclic heteroaromatic compounds
JP6506555B2 (en) 2011-10-19 2019-04-24 ファーマサイクリックス エルエルシー Use of Breton-type tyrosine kinase (Btk) inhibitors
US9550781B2 (en) 2011-11-14 2017-01-24 Centaurus Biopharma Co., Ltd. Kinase modulating compounds, compositions containing the same and use thereof

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002038561A1 (en) 2000-11-07 2002-05-16 Novartis Ag Indolylmaleimide derivatives as protein kinase c inhibitors
WO2003082859A1 (en) 2002-04-03 2003-10-09 Novartis Ag Indolylmaleimide derivatives
WO2004052359A1 (en) 2002-12-09 2004-06-24 The Board Of Regents Of The University Of Texas System Methods for selectively inhibiting janus tyrosine kinase 3 (jak3)
WO2004078163A2 (en) 2003-02-28 2004-09-16 Transform Pharmaceuticals, Inc. Pharmaceutical co-crystal compositions of drugs such as carbamazepine, celecoxib, olanzapine, itraconazole, topiramate, modafinil, 5-fluorouracil, hydrochlorothiazide, acetaminophen, aspirin, flurbiprofen, phenytoin and ibuprofen
WO2005000833A1 (en) 2003-05-19 2005-01-06 Irm, Llc Immunosuppressant compounds and compositions
WO2005066156A1 (en) 2004-01-12 2005-07-21 Cytopia Research Pty Ltd Selective kinase inhibitors
WO2007090752A1 (en) 2006-02-07 2007-08-16 F. Hoffmann-La Roche Ag Anthranilamide/2-amino-heteroarene carboxamide derivatives
WO2007145834A2 (en) 2006-06-08 2007-12-21 Amgen Inc. Benzamide derivatives and uses related thereto
US20090105209A1 (en) 2007-10-23 2009-04-23 Roche Palo Alto Llc BTK protein kinase inhibitors
WO2009077334A1 (en) 2007-12-14 2009-06-25 F. Hoffmann-La Roche Ag Novel imidazo[1,2-a]pyridine and imidazo[1,2-b]pyridazine derivatives
US20090306041A1 (en) 2008-02-05 2009-12-10 Roche Palo Alto Llc Inhibitors of Bruton's tyrosine kinase
WO2010000633A1 (en) 2008-07-02 2010-01-07 F. Hoffmann-La Roche Ag Novel phenylpyrazinones as kinase inhibitors

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY
"Remington's Pharmaceutical Sciences", 1990, MACK PRINTING COMPANY, pages: 1289 - 1329
DAVIS,R.E. ET AL., NATURE, vol. 463, 2010, pages 88 - 92
FATIH M UCKUN ET AL: "Bruton's tyrosine kinase as a molecular target in treatment of leukemias and lymphomas as well as inflammatory disorders and autoimmunity", EXPERT OPINION ON THERAPEUTIC PATENTS, INFORMA HEALTHCARE, GB, vol. 20, no. 11, 1 November 2010 (2010-11-01), pages 1457 - 1470, XP008157228, ISSN: 1354-3776, DOI: 10.1517/13543776.2010.517750 *
HATA,D. ET AL., J. EXP. MED., vol. 187, 1998, pages 1235 - 1247
J. MED. CHEM., vol. 52, no. 14, 2009, pages 4329 - 4337
J. MED. CHEM., vol. 52, no. 19, 2009, pages 5950 - 5966
STAHL; WERMUTH: "Handbook of Pharmaceutical Salts: Properties, Selection, and Use", 2002, WILEY-VCH
STEINBERG,B.J. ET AL., J. CLIN. INVEST, vol. 70, 1982, pages 587 - 597

Cited By (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9233111B2 (en) 2011-07-08 2016-01-12 Novartis Ag Pyrrolo pyrimidine derivatives
WO2013157021A1 (en) 2012-04-20 2013-10-24 Advinus Therapeutics Limited Bicyclic compounds, compositions and medicinal applications thereof
US9233983B2 (en) 2012-04-20 2016-01-12 Advinus Therapeutics Limited Substituted hetero-bicyclic compounds, compositions and medicinal applications thereof
AU2013250726B2 (en) * 2012-04-20 2017-01-05 Advinus Therapeutics Limited Substituted hetero-bicyclic compounds, compositions and medicinal applications thereof
WO2013157022A1 (en) 2012-04-20 2013-10-24 Advinus Therapeutics Limited Substituted hetero-bicyclic compounds, compositions and medicinal applications thereof
EP3550031A1 (en) 2012-07-24 2019-10-09 Pharmacyclics, LLC Mutations associated with resistance to inhibitors of bruton's tyrosine kinase (btk)
US9512084B2 (en) 2013-11-29 2016-12-06 Novartis Ag Amino pyrimidine derivatives
US11180460B2 (en) 2013-11-29 2021-11-23 Novartis Ag Amino pyrimidine derivatives
US11673868B2 (en) 2013-11-29 2023-06-13 Novartis Ag Amino pyrimidine derivatives
US10457647B2 (en) 2013-11-29 2019-10-29 Novartis Ag Amino pyrimidine derivatives
JP2020105181A (en) * 2013-12-02 2020-07-09 ファーマサイクリックス エルエルシー Methods of treating and preventing alloantibody driven chronic graft versus host disease
US11623921B2 (en) 2014-10-24 2023-04-11 Bristol-Myers Squibb Company Indole carboxamide compounds
EP3042903A1 (en) 2015-01-06 2016-07-13 Advinus Therapeutics Limited Substituted hetero-bicyclic compounds, compositions and medicinal applications thereof
US11407749B2 (en) 2015-10-19 2022-08-09 Incyte Corporation Heterocyclic compounds as immunomodulators
US11572366B2 (en) 2015-11-19 2023-02-07 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2017106634A1 (en) * 2015-12-17 2017-06-22 Incyte Corporation N-phenyl-pyridine-2-carboxamide derivatives and their use as pd-1/pd-l1 protein/protein interaction modulators
US11866435B2 (en) 2015-12-22 2024-01-09 Incyte Corporation Heterocyclic compounds as immunomodulators
US11535615B2 (en) 2015-12-22 2022-12-27 Incyte Corporation Heterocyclic compounds as immunomodulators
US11608337B2 (en) 2016-05-06 2023-03-21 Incyte Corporation Heterocyclic compounds as immunomodulators
US11673883B2 (en) 2016-05-26 2023-06-13 Incyte Corporation Heterocyclic compounds as immunomodulators
US11873309B2 (en) 2016-06-20 2024-01-16 Incyte Corporation Heterocyclic compounds as immunomodulators
US11718605B2 (en) 2016-07-14 2023-08-08 Incyte Corporation Heterocyclic compounds as immunomodulators
US11613536B2 (en) 2016-08-29 2023-03-28 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2018053437A1 (en) 2016-09-19 2018-03-22 Mei Pharma, Inc. Combination therapy
US11339149B2 (en) 2016-12-22 2022-05-24 Incyte Corporation Heterocyclic compounds as immunomodulators
US10308644B2 (en) 2016-12-22 2019-06-04 Incyte Corporation Heterocyclic compounds as immunomodulators
US11787793B2 (en) 2016-12-22 2023-10-17 Incyte Corporation Heterocyclic compounds as immunomodulators
US11465981B2 (en) 2016-12-22 2022-10-11 Incyte Corporation Heterocyclic compounds as immunomodulators
US10806785B2 (en) 2016-12-22 2020-10-20 Incyte Corporation Immunomodulator compounds and methods of use
US10793565B2 (en) 2016-12-22 2020-10-06 Incyte Corporation Heterocyclic compounds as immunomodulators
US11566026B2 (en) 2016-12-22 2023-01-31 Incyte Corporation Heterocyclic compounds as immunomodulators
US10800768B2 (en) 2016-12-22 2020-10-13 Incyte Corporation Heterocyclic compounds as immunomodulators
US11608334B2 (en) 2017-02-08 2023-03-21 The National Institutes of Pharmaceutical R&D Co., Ltd. Pyrrolo-aromatic heterocyclic compound, preparation method therefor, and medical use thereof
EP3556761A4 (en) * 2017-02-08 2019-12-25 The National Institutes Of Pharmaceutical Research Pyrrolo-aromatic heterocyclic compound, preparation method therefor, and medical use thereof
US11613543B2 (en) 2018-03-26 2023-03-28 Novartis Ag Substituted pyrrolo[2,3-d]pyrimidines as Bruton's Tyrosine Kinase inhibitors
EP4166557A1 (en) 2018-03-26 2023-04-19 Novartis AG Intermediates of n-(3-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)phenyl)benzamide derivatives
WO2019186343A1 (en) 2018-03-26 2019-10-03 Novartis Ag N-(3-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)phenyl)benzamide derivatives
WO2019186358A1 (en) 2018-03-26 2019-10-03 Novartis Ag 3-hydroxy-n-(3-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)phenyl)pyrrolidine-1-carboxamide derivatives
US11541056B2 (en) 2018-03-26 2023-01-03 Novartis Ag 3-hydroxy-N-(3-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)phenyl)pyrrolidine-1-carboxamide derivatives
US10669271B2 (en) 2018-03-30 2020-06-02 Incyte Corporation Heterocyclic compounds as immunomodulators
US11124511B2 (en) 2018-03-30 2021-09-21 Incyte Corporation Heterocyclic compounds as immunomodulators
US11414433B2 (en) 2018-05-11 2022-08-16 Incyte Corporation Heterocyclic compounds as immunomodulators
US10618916B2 (en) 2018-05-11 2020-04-14 Incyte Corporation Heterocyclic compounds as immunomodulators
US10906920B2 (en) 2018-05-11 2021-02-02 Incyte Corporation Heterocyclic compounds as immunomodulators
US11753406B2 (en) 2019-08-09 2023-09-12 Incyte Corporation Salts of a PD-1/PD-L1 inhibitor
US11401279B2 (en) 2019-09-30 2022-08-02 Incyte Corporation Pyrido[3,2-d]pyrimidine compounds as immunomodulators
US11866451B2 (en) 2019-11-11 2024-01-09 Incyte Corporation Salts and crystalline forms of a PD-1/PD-L1 inhibitor
US11780836B2 (en) 2020-11-06 2023-10-10 Incyte Corporation Process of preparing a PD-1/PD-L1 inhibitor
US11760756B2 (en) 2020-11-06 2023-09-19 Incyte Corporation Crystalline form of a PD-1/PD-L1 inhibitor
US11866434B2 (en) 2020-11-06 2024-01-09 Incyte Corporation Process for making a PD-1/PD-L1 inhibitor and salts and crystalline forms thereof
WO2022189859A1 (en) 2021-03-12 2022-09-15 Novartis Ag Fatty acid-bifunctional degrader conjugates and their methods of use

Also Published As

Publication number Publication date
AU2012282229B2 (en) 2015-05-07
EP2729466B1 (en) 2015-08-19
EA201490229A1 (en) 2014-05-30
US20140243306A1 (en) 2014-08-28
CN103732596B (en) 2016-06-01
AU2012282229A1 (en) 2014-01-16
ES2548414T3 (en) 2015-10-16
CN103732596A (en) 2014-04-16
BR112014000314A2 (en) 2017-01-10
CA2841111A1 (en) 2013-01-17
US9233111B2 (en) 2016-01-12
JP6145451B2 (en) 2017-06-14
EP2729466A1 (en) 2014-05-14
KR20140058543A (en) 2014-05-14
JP2014520793A (en) 2014-08-25
MX2014000338A (en) 2014-05-01

Similar Documents

Publication Publication Date Title
EP2729466B1 (en) Novel pyrrolo pyrimidine derivatives
US11673868B2 (en) Amino pyrimidine derivatives
EP2897961B1 (en) Dihydropyrrolidino-pyrimidines as kinase inhibitors
US20150216867A1 (en) Compounds for use in gastric complication
TW201425313A (en) Novel pyrrolo pyrimidine derivatives

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 12761655

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2012761655

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2014517980

Country of ref document: JP

Kind code of ref document: A

Ref document number: 2841111

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: MX/A/2014/000338

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2012282229

Country of ref document: AU

Date of ref document: 20120707

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20147002986

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 201490229

Country of ref document: EA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112014000314

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: 14130536

Country of ref document: US

ENP Entry into the national phase

Ref document number: 112014000314

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20140107